Language selection

Search

Patent 2751321 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 2751321
(54) English Title: METHODS OF ADMINISTERING (4AR, 1OAR)-1-N-PROPYL-1,2,3,4A,5,10,10A-OCTAHYDROBENZO [G] QUINOLINE-6,7-DIOL AND RELATED COMPOUNDS ACROSS THE ORAL MUCOSA, THE NASAL MUCOSA OR THE SKIN AND PHARMACEUTICAL COMPOSITIONS THEREOF
(54) French Title: METHODES D'ADMINISTRATION DE (4AR, 1OAR)-1-N-PROPYL-1,2,3,4A,5,10, 10A-OCTAHYDROBENZO [G] QUINOLEINE-6,7-DIOL ET DE COMPOSES APPARENTES A TRAVERS_LA MUQUEUSE BUCCALE, LA MUQUEUSE N ASALE OU LA PEAU ET LEURS COMPOSITIONS PHARMACEUTIQUES
Status: Dead
Bibliographic Data
(51) International Patent Classification (IPC):
  • A61K 9/20 (2006.01)
  • A61K 9/68 (2006.01)
  • A61K 31/473 (2006.01)
  • A61P 25/16 (2006.01)
(72) Inventors :
  • WIKSTROEM, HAKAN (Sweden)
  • JOERGENSEN, MORTEN (Denmark)
  • MOERK, NIELS (Denmark)
  • LARSEN, JENNIFER (Denmark)
  • BANG-ANDERSEN, BENNY (Denmark)
  • SAGER, THOMAS NIKOLAJ (Denmark)
  • PUESCHL, ASK (Denmark)
  • TORUP, LARS (Denmark)
(73) Owners :
  • H. LUNDBECK A/S (Denmark)
(71) Applicants :
  • H. LUNDBECK A/S (Denmark)
(74) Agent: GOUDREAU GAGE DUBUC
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2010-02-26
(87) Open to Public Inspection: 2010-09-02
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/DK2010/050050
(87) International Publication Number: WO2010/097091
(85) National Entry: 2011-08-02

(30) Application Priority Data:
Application No. Country/Territory Date
PA 200900274 Denmark 2009-02-27
61/155,933 United States of America 2009-02-27
PA 200900279 Denmark 2009-02-27
61/155,942 United States of America 2009-02-27
PA 200900282 Denmark 2009-02-27
61/155,957 United States of America 2009-02-27

Abstracts

English Abstract





Disclosed are pharmaceutical compositions and methods for the administration
of (4aR,10aR)-1- n-propyl
-1,2,3,4,4a,5,10,10a-octahydro-benzo[g]quinoline-6,7-diol or a
pharmaceutically acceptable salt thereof and related compounds
for the treatment of neurological disorder such as Parkinson's disease and
restless leg syndrome.


French Abstract

La présente invention concerne des compositions pharmaceutiques et des méthodes d'administration de (4aR,10aR)-1-n-propyl-1,2,3,4,4a,5,10,10a-octahydro-benzo[g]quinoléine-6,7-diol ou d'un sel pharmaceutiquement acceptable de ce composé et de composés apparentés aux fins du traitement d'affections neurologiques telles que la maladie de Parkinson et le syndrome des jambes sans repos.

Claims

Note: Claims are shown in the official language in which they were submitted.





74



CLAIMS

1. A pharmaceutical composition for delivery across the oral mucosa, nasal
mucosa or skin
comprising (4aR,10aR)-1-n-propyl-1,2,3,4,4a,5,10,10a-octahydro-
benzo[g]quinoline-6,7-
diol or a pharmaceutically acceptable salt thereof, and a pharmaceutically
acceptable
carrier.


2. Use of a pharmaceutical composition for delivery across the oral mucosa,
nasal mucosa
or skin comprising (4aR,10aR)-1-n-propyl-1,2,3,4,4a,5,10,10a-octahydro-
benzo[g]quinoline-6,7-diol or a pharmaceutically acceptable salt thereof, in
the
preparation of a medicament for the treatment of Parkinson's disease.


3. The pharmaceutical composition of claim 1 for delivery across the oral
mucosa
comprising (4aR,10aR)-1-n-propyl-1,2,3,4,4a,5,10,10a-octahydro-
benzo[g]quinoline-6,7-
diol or a pharmaceutically acceptable salt thereof, and a pharmaceutically
acceptable
carrier.


4. The use of claim 2 for delivery across the oral mucosa comprising
(4aR,10aR)-1-n-
propyl-1,2,3,4,4a,5,10,10a-octahydro-benzo[g]quinoline-6,7-diol or a
pharmaceutically
acceptable salt thereof, in the preparation of a medicament for the treatment
of
Parkinson's disease.


5. The pharmaceutical composition of claim 3 wherein the delivery across the
oral mucosa
occurs through oral buccal route, sublingual route or through the lips.


6. The pharmaceutical composition of claim 3 or 5, wherein said composition
further
comprises a penetration enhancer.


7. The pharmaceutical composition of anyone of claims 3, 5 and 6, wherein said

composition is a tablet, lozenge, chewing gum or lipstick.


8. A pharmaceutical composition for intranasal administration comprising
(4aR,10aR)-1-n-
propyl-1,2,3,4,4a,5,10,10a-octahydro-benzo[g]quinoline-6,7-diol or a
pharmaceutically
acceptable salt thereof, and a pharmaceutically acceptable carrier.





75



9. Use of a pharmaceutical composition for intranasal delivery comprising
(4aR,10aR)-1-n-
propyl-1,2,3,4,4a,5,10,10a-octahydro-benzo[g]quinoline-6,7-diol or a
pharmaceutically
acceptable salt thereof, in the preparation of a medicament for the treatment
of
Parkinson's disease.


10. The pharmaceutical composition of claim 8 further comprising a permeation
enhancer.


11. The pharmaceutical composition of claim 8 or 10 further comprising one or
more
adhesive, binder, lubricant, glidant, disintegrant or mixture thereof.


12. A pharmaceutical composition for transdermal delivery comprising
(4aR,10aR)-1-n-
propyl-1,2,3,4,4a,5,10,10a-octahydro-benzo[g]quinoline-6,7-diol or a
pharmaceutically
acceptable salt thereof, and a pharmaceutically acceptable carrier.


13. Use of a pharmaceutical composition for transdermal delivery comprising
(4aR,10aR)-1-
n-propyl-1,2,3,4,4a,5,10,10a-octahydro-benzo[g]quinoline-6,7-diol or a
pharmaceutically
acceptable salt thereof, in the preparation of a medicament for the treatment
of
Parkinson's disease.


14. The pharmaceutical composition of claim 12 further characterized as patch,
a semisolid
dosage form, a gel, lotion or crème.


15. The pharmaceutical composition of claim 12 or 14, further comprising a
permeation
enhancer, one or more adhesive, binder, lubricant, glidant, disintegrant or a
mixture
thereof.


16. A pharmaceutical composition for delivery across the oral mucosa, nasal
mucosa or skin
comprising a compound selected from Formula 1a, 1b or 1c:
Image




76



wherein each R x, R y, and R z is independently C1-6 alkanoyl,
cycloalkylalkyl, phenylacetyl
or benzoyl, or a pharmaceutically acceptable salt thereof, and a
pharmaceutically
acceptable carrier.

Description

Note: Descriptions are shown in the official language in which they were submitted.



CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
METHODS OF ADMINISTERING (4AR,
10AR)-1-N-PROPYL-1,2,3,4A,5,10, 10A-OCTAHYDROBENZO [G] QUINOLINE-6,7-DIOL AND
RELATED COMPOUNDS ACROSS THE ORAL MUCOSA, THE NASAL MUCOSA OR THE SKIN AND
PHARMACEUTICAL COMPOSITIONS THEREOF

FIELD OF THE INVENTION
The present invention relates to methods of administering (4aR,l0aR)-1-n-
propyl-
1,2,3,4,4a,5,10,10a-octahydro-benzo[g]quinoline-6,7-diol for the treatment of
neurological
disorders and pharmaceutical compositions thereof.

BACKGROUND ART
The use of dopamine-replacing agents in the symptomatic treatment of
Parkinson's disease (PD)
has undoubtedly been successful in increasing the quality of life of patients.
L-DOPA, which has
been used for many years and remains the gold standard for treatment of PD,
alleviates motor
symptoms of PD characterized by the slowness of movement (bradykinesia),
rigidity and/or
tremor. It is understood that L-DOPA acts as a prodrug which is bio-
metabolized into dopamine
(DA). DA in turn activates dopamine receptors in the brain which fall into two
classes: Dl and
D2 receptors. Dl receptors can be divided into D1 and D5 receptors while D2
receptors can be
divided into D2, D3, and D4 receptors. However, dopamine-replacement therapy
does have
limitations, especially following long-term treatment.

PD afflicted patients may cycle between "on" periods in which normal
functioning is attained and
"off' periods in which they are severely parkinsonian. Additionally, as a
consequence they may
experience profound disability despite the fact that L-DOPA remains an
effective anti-Parkinson
agent throughout the course of the disease (Obeso, JA, et at. Neurology 2000,
55, S13-23). It is
worth noting that DA agonists do cause less dyskinesia than L-DOPA but this is
of limited value
to PD patients with dyskinesias because many of them have moderate-to-severe
PD and often
they need the efficacy of L-DOPA.

Anti-Parkinson agents that mimic the action of DA have been shown to be
effective in treating
PD. Selective D2-agonists such as Pramipexole are effective but lack efficacy
in late PD and
eventually need complementation or replacement with L-DOPA. Apomorphine is a
catecholamine anti-Parkinson's agent that acts as a potent D1/D2 agonist. In
particular, this drug
is useful as a rescue during the "off' periods of severely disabled patients
who have received
chronic L-DOPA treatment. However, due to its poor oral bioavailability and
high first-pass
effect, apomorphine is limited in its clinical application. To overcome the
high first pass effect


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
2
and poor oral bioavailability, apomorphine must be administered
subcutaneously. Generally, the
poor oral bioavailability of catecholamines has prevented their clinical use
as orally administered
drugs.

Apart from PD, other diseases in which an increase in dopaminergic turnover
may be beneficial
include treating depression and for the improvement of mental functions
including various
aspects of cognition. Dopaminergic turnover can have a positive effect on the
treatment of
obesity as an anorectic agent. It can improve minimal brain dysfunction (MBD),
narcolepsy, and
potentially the negative, the positive as well as the cognitive symptoms of
schizophrenia. Restless
leg syndrome (RLS) and periodic limb movement disorder (PLMD) are alternative
indications,
which are clinically treated with DA agonists.

In addition, impotence and erectile dysfunction are also likely to be improved
by treatment with
DA agonists. Thus, improvement of sexual functions in both women and men is
another possible
indication for treatment with DA agonists since erectile dysfunction
(impotence in men) and
sexual stimulation in e.g. menopausal women (stimulation of vaginal
lubrication and erection of
clitoris) potentially can be achieved via DA receptor stimulation. In this
context, it is noteworthy
that apomorphine when given sublingually is used clinically to improve
erectile dysfunction.

Clinical studies of L-DOPA and the D2 agonist Pramipexole as therapies in
Huntington's disease
have shown promising results; thus treatment of Huntington's disease is
another potential
application of the compounds of the invention. DA is involved in regulation of
the cardiovascular
and renal systems, and accordingly, renal failure and hypertension can be
considered alternative
indications for the compounds of the invention.
Despite the long-standing interest in the field, there is evidently an unmet
need for developing
efficient and active drugs for the treatment of PD. A mixed D1/D2 agonist
giving continuous
dopaminergic stimulation may fulfil such unmet needs. To this end, (4aR,lOaR)-
1-n-propyl-
1,2,3,4,4a,5,10,1Oa-octahydro-benzo[g]quinoline-6,7-diol [herein referred to
as Compound 10]
has been identified as a potent D1/D2 agonist which shows potential to treat
PD. However, as
previously mentioned, the poor oral bioavailability of catecholamines has
prevented their clinical
use as orally administered drugs.


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
3
Alternatively, the oral mucosal delivery of drugs utilizes primarily the
sublingual and buccal
mucosas as absorption sites, although the whole oral cavity can be considered
for both mucosal
(local effect) and trans-mucosal (systemic effect) absorption of drugs. Owing
to the ease of
administration, the oral cavity is an attractive site for delivery of drugs.
Furthermore, the oral
cavity has reduced enzymatic activity as compared to the intestinal, rectal,
and nasal mucosas,
which may lead to an improved absorption and a reduced irritation at this site
of absorption. The
oral cavity is less sensitive to damage and irritation than the nasal
epithelium.

The oral mucosa provides a protective coating for underlying tissues while
acting as a barrier to
microorganisms and as a control to the passage of substances through the oral
cavity. In humans,
the buccal membranes consist of keratinized and nonkeratinized striated
epithelium. Many
factors, including partition characteristics, degree of ionization, and
molecular size, influence the
transport of drugs across the membrane. However, many drugs do not pass
through the buccal
membranes in sufficient amounts to be useful.

In general, the sublingual route is preferred for disorders requiring acute
drug delivery whereas
the buccal route is often utilized in cases where a prolonged drug delivery is
desirable.
Furthermore, a sublingual or buccal drug formulation offers an attractive
alternative for patients
e.g. patients suffering from Parkinson's disease having difficulties
swallowing conventional oral
drug formulations such as tablets or capsules. For reviews on buccal drug
delivery, see: Shojaei,
J. of Pharmacy & Pharm. Sci., 1998, 1, 15; Rossi et al, Drug Discovery Today
2005, 2, 1, 59; and
Pather et at. Expert Opinion on Drug Delivery 2008, 5, 531. The sublingual
route usually
produces a faster onset of action than traditional orally administered tablets
and the portion
absorbed through the sublingual blood vessels bypasses the hepatic first pass
metabolic processes
(Motwani et at., Clin. Pharm. 1991, 21, 83-94; and Ishikawa et at., Chem.
Pharm. Bull. 2001, 49,
230-232).

Due to high buccal vascularity, buccally delivered drugs can gain direct
access to the systemic
circulation and are not subject to first-pass hepatic metabolism. In addition,
therapeutic agents
administered via the buccal route are not exposed to the environment of the
gastrointestinal tract
(Mitra et at., Encyclopedia of Pharm. Tech. 2002, 2081-2095). Further, the
buccal mucosa has
low enzymatic activity relative to the nasal and rectal routes. Thus, the
potential for drug
inactivation due to biochemical degradation is less rapid and extensive than
other administration
routes (de Varies et at., Crit. Rev. Ther. Drug Carr. Syst. 1999, 8, 271-303).


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
4
Since the oral mucosa is renewed relatively fast, discoloration of the oral
cavity is minimized
with buccal delivery as compared to other modes of delivery. Buccal delivery
is also
advantageous over other modes of delivery. For example, local skin irritations
are observed with
the transdermal delivery of catecholamines. Further, irritation at the
injection site and
precipitation of decomposed apomorphine are sometimes associated with its
intermittent
subcutaneous administration as well as with delivery via continuous infusion.

To this end, the inventors have discovered methods to administer (4aR,10aR)-l-
n-propyl-
1,2,3,4,4a,5,10,10a-octahydro-benzo[g]quinoline-6,7-diol and related compounds
via oral
mucosa delivery. This has been achieved by the development of novel
pharmaceutical
compositions of said compounds for buccal administration in the treatment of
Parkinson's disease
as well as the other conditions disclosed in this application. Accordingly,
the present invention
provides pharmaceutical compositions for buccal administration comprising one
of the
compounds of the invention, or a pharmaceutically acceptable salt, and a
pharmaceutically
acceptable carrier.
Separately, the nasal mucosa offers an alternative to oral and parenteral
administration; intranasal
administration is a practical way to achieve the therapeutic effect of many
medications.
Advantages of this method are that drugs can be administered readily and
simply, and either a
localized or a systemic effect can be achieved. In nasal administration, the
biologically active
substance must be applied to the nasal mucosa in such a condition that it is
able to penetrate or be
absorbed through the mucosa. The extensive network of blood capillaries under
the nasal mucosa
is particularly suited to provide a rapid and effective systemic absorption of
drugs. Moreover, the
nasal epithelial membrane consists of practically a single layer of epithelial
cells (pseudostratified
epithelium) and may be more suited for drug administration than other mucosal
surfaces having
squamous epithelial layers, such as the mouth, vagina, etc.

Further, the intranasal administration of drugs that exert their effect in the
brain may have the
advantage in that the blood-brain-barrier (BBB) may be a less of a hurdle for
the drug than if the
drug had to traverse the BBB through the `normal' blood stream. The onset of
action may also
be significantly faster for the intranasal administration of CNS based drugs
than by other routes
of administration.


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
The inventors have discovered methods to administer (4aR,l0aR)-1-n-propyl-
1,2,3,4,4a,5,10,10a-octahydro-benzo[g]quinoline-6,7-diol and related compounds
via intranasal
administration. This has been achieved by the development of novel
pharmaceutical
compositions of said compounds for intranasal administration in the treatment
of Parkinson's
5 disease as well as the other conditions disclosed in this application.
Accordingly, the present
invention provides pharmaceutical compositions for intranasal administration
comprising one of
the compounds of the invention, or a pharmaceutically acceptable salt, and a
pharmaceutically
acceptable carrier.

Moreover, delivering pharmaceutical agents into the systemic circulation
through the skin is seen
as a desirable route of administration while providing several other
advantages over oral
administration. For example, bypassing the gastrointestinal (GI) tract would
obviate the GI
irritation that frequently occurs and avoid partial first-pass inactivation by
the liver. Further,
steady absorption of drug over hours or days can be preferable to the blood
level spikes and
troughs produced by oral dosage forms. Additionally, patients often forget to
take their medicine
and even the most faithfully compliant get tired of swallowing pills,
especially if they must take
several each day. The transdermal route can also be more effective than the
oral route in that it
can provide for relatively faster or slower (extended) absorption and onset of
therapeutic action.
Transdermal delivery also poses inherent challenges, in part because of the
nature of skin. Skin is
essentially a thick membrane that protects the body by acting as a barrier.
Consequently, the
movement of drugs or any external agent through the skin is a complex process.
The structure of
skin includes the relatively thin epidermis, or outer layer, and a thicker
inner layer called the
dermis. For a drug to penetrate unbroken skin, it must first move into and
through the stratum
comeum, which is the outer layer of the epidermis. Then the drug must
penetrate the viable
epidermis, papillary dermis, and capillary walls to enter the blood stream or
lymph channels.
Each tissue features a different resistance to penetration, but the stratum
corneum is the strongest
barrier to the absorption of transdermal and topical drugs. The tightly packed
cells of the stratum
corneum are filled with keratin. The keratinization and density of the cells
may be responsible for
skin's impermeability to certain drugs.

In recent years, advances in transdermal delivery include the formulation of
permeation
enhancers (skin penetration enhancing agents). Permeation enhancers often are
lipophilic
chemicals that readily move into the stratum comeum and enhance the movement
of drugs


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
6
through the skin. Non-chemical modes also have emerged to improve transdermal
delivery; these
include ultrasound, iontophoresis, and electroporation.

The inventors have discovered methods to administer (4aR,lOaR)-l-n-propyl-
1,2,3,4,4a,5,10,1Oa-octahydro-benzo[g]quinoline-6,7-diol and related compounds
via transdermal
delivery. This has been achieved by the development of novel pharmaceutical
compositions of
said compounds for transdermal administration in the treatment of Parkinson's
disease as well as
the other conditions disclosed in this application. Accordingly, the present
invention provides
pharmaceutical compositions for transdermal administration comprising one of
the compounds of
the invention, or a pharmaceutically acceptable salt, and a pharmaceutically
acceptable carrier.
SUMMARY OF THE INVENTION
The present invention relates a pharmaceutical composition for delivery across
the oral mucosa,
nasal mucosa or skin comprising (4aR,1OaR)-l-n-propyl-1,2,3,4,4a,5,10,1Oa-
octahydro-
benzo[g]quinoline-6,7-diol or a pharmaceutically acceptable salt thereof, and
a pharmaceutically
acceptable carrier.

Another aspect relates to a use of a pharmaceutical composition for delivery
across the oral
mucosa, nasal mucosa or skin comprising (4aR,1OaR)-1-n-propyl-
1,2,3,4,4a,5,10,1Oa-octahydro-
benzo[g]quinoline-6,7-diol or a pharmaceutically acceptable salt thereof, in
the preparation of a
medicament for the treatment of Parkinson's disease.

Further, aspects of the present invention relate to a pharmaceutical
composition for delivery
across the oral mucosa comprising (4aR,1OaR)-l-n-propyl-1,2,3,4,4a,5,10,1Oa-
octahydro-
benzo[g]quinoline-6,7-diol or a pharmaceutically acceptable salt thereof, and
a pharmaceutically
acceptable carrier. A separate aspect is directed to a pharmaceutical
composition for delivery
across the oral mucosa comprising racemic trans- l-n-propyl-
1,2,3,4,4a,5,10,1Oa-octahydro-
benzo[g]quinoline-6,7-diol or a pharmaceutically acceptable salt thereof, and
a pharmaceutically
acceptable carrier.

Another aspect relates to a method for the delivery across the oral mucosa of
the (4aR,lOaR)
enantiomer or the racemic trans isomer of 1-n-propyl-1,2,3,4,4a,5,10,10a-
octahydro-
benzo[g]quinoline-6,7-diol or a pharmaceutically acceptable salt thereof.
Separately, an aspect of
the invention relates to the use of a pharmaceutical composition for delivery
across the oral


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
7
mucosa comprising a therapeutically effective amount of the (4aR,1 OaR)
enantiomer or the
racemic trans isomer of 1-n-propyl-1,2,3,4,4a,5,10,10a-octahydro-
benzo[g]quinoline-6,7-diol or
a pharmaceutically acceptable salt thereof, in the preparation of a medicament
for treating a
neurological disorder. In one aspect, the neurological disorder is Parkinson's
disease.
A separate concern of the invention is directed to a method of treating a
neurological disorder
comprising administering a pharmaceutical composition for delivery across the
oral mucosa of a
therapeutically effective amount of the (4aR,1 OaR) enantiomer or the racemic
trans isomer of 1-
n-propyl-1,2,3,4,4a,5, 10, 10a-octahydro-benzo[g]quinoline-6,7-diol or a
pharmaceutically
acceptable salt thereof. In one aspect, the neurological disorder is
Parkinson's disease.

Yet another aspect of the present invention relates to a pharmaceutical
composition for intranasal
administration comprising (4aR,1OaR)-1-n-propyl-1,2,3,4,4a,5,10,10a-octahydro-
benzo[g]quinoline-6,7-diol or a pharmaceutically acceptable salt thereof, and
a pharmaceutically
acceptable carrier. A separate aspect is directed to a pharmaceutical
composition for intranasal
administration comprising racemic trans- l-n-propyl-1,2,3,4,4a,5,10,10a-
octahydro-
benzo[g]quinoline-6,7-diol or a pharmaceutically acceptable salt thereof, and
a pharmaceutically
acceptable carrier.

Another aspect relates to a method for the intranasal delivery of the (4aR, l
OaR) enantiomer or the
racemic trans isomer of 1-n-propyl-1,2,3,4,4a,5,10,10a-octahydro-
benzo[g]quinoline-6,7-diol or
a pharmaceutically acceptable salt thereof Separately, an aspect of the
invention relates to the
use of a pharmaceutical composition for intranasal administration comprising a
therapeutically
effective amount of the (4aR,1OaR) enantiomer or racemic trans isomer of 1-n-
propyl-
1,2,3,4,4a,5,10,10a-octahydro-benzo[g]quinoline-6,7-diol or a pharmaceutically
acceptable salt
thereof, in the preparation of a medicament for treating a neurological
disorder. In one aspect,
the neurological disorder is Parkinson's disease.

A separate concern of the invention is directed to a method of treating a
neurological disorder
comprising administering a pharmaceutical composition for intranasal
administration of a
therapeutically effective amount of the (4aR,1 OaR) enantiomer or the racemic
trans isomer of 1-
n-propyl- 1,2,3,4,4a,5,10,10a-octahydro-benzo[g]quinoline-6,7-diol or a
pharmaceutically
acceptable salt thereof. In one aspect, the neurological disorder is
Parkinson's disease.


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
8
One aspect of the present invention relates to a pharmaceutical composition
for transdermal
delivery comprising (4aR,1OaR)-l-n-propyl-1,2,3,4,4a,5,10,1 Oa-octahydro-
benzo[g]quinoline-
6,7-diol or a pharmaceutically acceptable salt thereof, and a pharmaceutically
acceptable carrier.
A separate aspect is directed to a pharmaceutical composition for transdermal
delivery
comprising racemic trans- l-n-propyl-1,2,3,4,4a,5, 10,1Oa-octahydro-
benzo[g]quinoline-6,7-diol
or a pharmaceutically acceptable salt thereof, and a pharmaceutically
acceptable carrier.

Another aspect relates to a method for a pharmaceutical composition for
transdermal delivery
comprising the (4aR,lOaR) enantiomer or the racemic trans isomer of 1-n-propyl-

1,2,3,4,4a,5,10,10a-octahydro-benzo[g]quinoline-6,7-diol or a pharmaceutically
acceptable salt
thereof Separately, an aspect of the invention relates to the use of a
pharmaceutical composition
for transdermal delivery comprising a therapeutically effective amount of the
(4aR,1OaR)
enantiomer or the racemic trans isomer of 1-n-propyl-1,2,3,4,4a,5,10,10a-
octahydro-
benzo[g]quinoline-6,7-diol or a pharmaceutically acceptable salt thereof, in
the preparation of a
medicament for treating a neurological disorder. In one aspect, the
neurological disorder is
Parkinson's disease.

A separate concern of the invention relates to a method of treating a
neurological disorder
comprising administering a pharmaceutical composition for transdermal delivery
of a
therapeutically effective amount of the (4aR,1 OaR) enantiomer or the racemic
trans isomer of 1-
n-propyl- 1,2,3,4,4a,5,10,10a-octahydro-benzo[g]quinoline-6,7-diol or a
pharmaceutically
acceptable salt thereof In one aspect, the neurological disorder is
Parkinson's disease.

Yet another aspect relates to a pharmaceutical composition for delivery across
the oral mucosa,
nasal mucosa or skin comprising a compound selected from Formula 1 a, l b or 1
c:

R 0 O114, O HO
0yR, OH OyRz
O O
Formula la Formula lb Formula lc

wherein each RX, Ry, and Rz is independently C1.6 alkanoyl, cycloalkylalkyl,
phenylacetyl or
benzoyl, or a pharmaceutically acceptable salt thereof, and a pharmaceutically
acceptable carrier.


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
9
One aspect of the invention is directed to a ratio from about 0:1 to about 1:0
of a mixture of the
asymmetric diesters of Formula la wherein Rx # Ry. A separate aspect of the
invention relates to
a ratio from about 0:1 to about 1:0 of a mixture of the mono-esters of
Formulas Ib and Ic.

Separate aspects of the invention are directed to the uses and methods of the
pharmaceutical
compositions described above for the treatment of Parkinson's disease.

DETAILED DESCRIPTION
The compounds of the present invention contain two chiral centers (denoted
with * in the below
formula)

\ N
HO'
OH
Example I

The compounds of the invention can exist in two different diastereomeric
forms, the cis- and
trans-isomers, both of which can exist in two enantiomeric forms. The present
invention relates
only to the trans racemate and the (4aR, l OaR)-enantiomer.
racemates enantiomers
\ I \ I \ ~r.N
HO HO HO
OH OH OH
cis diastereomers cis racemate of formula I (4aR, lOaS)-enantiomer (4aS, lOaR)-
enantiomer
trans diastereomers

I \ ~A~.N
\ ) HO' I \ ) HO'
/
.
HO . "b
OH OH OH
trans racemate of formula I (4aR, lOaR)-enantiomer (4aS, lOaS)-enantiomer

As previously indicated, the present invention is based on the discovery that
(4aR,1OaR)-l-n-
propyl- 1,2,3,4,4a,5,10,10a-octahydro-benzo[g]quinoline-6,7-diol (herein
referred to as
"Compound 10") is a potent Dl / D2 agonist which is bioavailable via delivery
through the oral


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
mucosa. The invention is explained in greater detail below but this
description is not intended to
be a detailed catalog of all the different ways in which the invention may be
implemented, or all
the features that may be added to the instant invention.

5 Racemic trans- l-n-propyl-1,2,3,4,4a,5,10,10a-octahydro-benzo[g]quinoline-
6,7-diol is a 1:1
mixture of (4aR,1OaR)-l-n-propyl-1,2,3,4,4a,5,10,10a-octahydro-
benzo[g]quinoline-6,7-diol and
(4aS, l OaS)-l -n-propyl-1,2,3,4,4a,5,10,10a-octahydro-benzo[g]quinoline-6,7-
diol.

"Related compounds of (4aR,1OaR)-1-n-propyl-1,2,3,4,4a,5,10,10a-octahydro-
10 benzo[g]quinoline-6,7-diol" refer to racemic trans- l-n-propyl-
1,2,3,4,4a,5,10,10a-octahydro-
benzo[g]quinoline-6,7-diols and the symmetric, asymmetric and mono-esters of
Formulas Ia, Ib
and Ic. Both the racemic trans isomer and the (4aR,10aR)-enantiomer of
Formulas Ia, Ib and Ic
fall within the scope of the invention.

As used herein, "C1-6 alkanoyl" refers to a straight-chain or branched-chain
alkanoyl group
containing from one to six carbon atoms, examples of which include a formyl
group, an acetyl
group, a pivaloyl group, and the like.

"Cycloalkylalkyl" refers to a saturated carbocyclic ring attached to a
terminal end of an a straight-
chain or branched-chain alkylene linker containing one to three carbon atoms,
examples of which
include a cyclopropylmethyl group, a cyclobutylethyl group, a
cyclopentylpropyl group, and the
like.

As used herein, "active ingredient" or the "compound of the invention" refers
to a compound
selected from the group consisting of (4aR,iOaR)-l-n-propyl-
1,2,3,4,4a,5,10,10a-octahydro-
benzo[g]quinoline-6,7-diol; racemic trans 1-n-propyl-1,2,3,4,4a,5,10,10a-
octahydro-
benzo[g]quinoline-6,7-diol; or a compound of Formulas Ia, lb or Ic. Both the
racemic trans
isomer and the (4aR,10aR)-enantiomer of Formulas Ia, Ib and Ic fall within the
scope of the
invention.
A. Administration across the Oral Mucosa
As used herein, the "oral mucosal" membranes of the buccal cavity encompass
the following five
regions: the buccal mucosa (cheeks), the floor of the mouth (sublingual), the
gums (gingiva), the
palatal mucosa, and the lining of the lips.


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
11
The pharmaceutical compositions described herein may contain permeation
enhancers because
the buccal cavity is a poor absorptive site of the alimentary tract. The
buccal cavity lacks the
typical villus-type of absorptive membrane of the intestine. Further, unlike
the intestine, the
junction between epithelial cells are tight. For a substance to be absorbed
through the mucosal
membrane of the buccal cavity, it should be presented in a lipophilic form.

The delivery systems in accordance with the present invention may be used in
conjunction with
permeation/absorption enhancers known in the art. Suitable examples include:
anionic
surfactants (e.g. sodium lauryl sulfate, sodium laureate); cationic
surfactants (e.g. cetylpyridinium
chloride); nonionic surfactants (e.g. Polysorbate-80); bile salts (e.g. sodium
glycodeoxycholate,
sodium glycocholate, sodium taurodeoxycholate, sodium taurocholate);
Polysaccharides (e.g.
Chitosan); Synthetic polymers (e.g. Carbopol, Carbomer); Fatty acids (e.g.
Oleic acid, Caprylic
acid); Chelators (e.g. = Ethylenediaminetetraacetic acid, Sodium citrate); and
Cyclodextrins: a,
0, y cyclodextrins. For a general review and insights on mechanism of action
of absorption
(permeation) enhancers for buccal application such as increasing the fluidity
of the cell
membrane, extracting inter/intracellular lipids, altering cellular proteins or
altering surface mucin
it is referred to Senel, J. Control. Res., 2001, 72:133-144.

Antioxidants
The buccal compositions can also include one or more antioxidants.
Representative antioxidants
include quaternary ammonium salts such as lauralkonium chloride, benzalkonium
chloride,
benzododecinium chloride, cetyl pyridium chloride, cetrimide, domiphen
bromide; alcohols such
as benzyl alcohol, chlorobutanol, o-cresol, phenyl ethyl alcohol; organic
acids or salts thereof
such as ascorbic acid, benzoic acid, sodium benzoate, sodium ascorbate,
potassium sorbate,
parabens; or complex forming agents such as EDTA.

Other Excipients
The carriers and excipients include ion-exchange microspheres which carry
suitable anionic
groups such as carboxylic acid residues, carboxymethyl groups, sulphopropyl
groups and
methylsulphonate groups. Ion-exchange resins, such as cation exchangers, can
also be used.
Chitosan, which is partially deacetylated chitin, or poly-N-acetyl-D-
glucosamine, or a
pharmaceutically acceptable salt thereof such as hydrochloride, lactate,
glutamate, maleate,
acetate, formate, propionate, maleate, malonate, adipate, or succinate.
Suitable other ingredients
for use as non-ion-exchange microspheres include starch, gelatin, collagen and
albumin.


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
12
pH Adjustment
Excipients to adjust the tonicity of the composition may be added such as
sodium chloride,
glucose, dextrose, mannitol, sorbitol, lactose, and the like. Acidic or basic
buffers can also be
added to the oral mucosal composition to control the pH. Low pH may be
preferable in the
instant case.

The compound of the invention as a pharmaceutical composition, may be
administered in any
suitable way in the oral cavity, and the compound may be presented in any
suitable dosage form
for such administration, e.g. in form of simple solutions or dispersions,
simple tablets, matrix
tablets, capsules, powders, syrups, dissolvable films, patches, lipophilic
gels. In one embodiment,
the compound of the invention is administered in the form of a solid
pharmaceutical entity,
suitably as a tablet or a capsule. In another particular embodiment, the
compound of the invention
is administered in the form of a dissolvable film.
In the case of oral mucosal administration of the compound of the invention,
conventional dosage
forms may not be able to assure therapeutic drug levels in because of
physiological removal
mechanism of the oral cavity (washing effect of saliva and mechanical stress),
which remove the
drug formulation away from the oral mucosa, resulting in too short exposure
time and
unpredictable absorption. To obtain the desired therapeutic action it may
therefore be necessary
to prolong and improve the contact between the compound of the invention and
the mucosa. To
fulfill the therapeutic requirement, formulations designed for sublingual or
buccal administration
may therefore contain mucoadhesive agents to maintain an intimate and
prolonged contact of the
formulation with the absorption site; penetration enhancers, to improve drug
permeation across
the mucosa; and enzyme inhibitors to eventually protect the drug from
degradation by means of
oral mucosal enzymes.

In one embodiment, the delivery across the oral mucosa occurs through buccal
route. In another
embodiment, the delivery across the oral mucosa occurs through the sublingual
route. In another
embodiment, the delivery across the oral mucosa occurs through the lips. In
one embodiment,
the pharmaceutical composition is a liquid solution. In one embodiment, the
pharmaceutical
composition is a gel. In yet another embodiment, the composition further
comprises a
penetration enhancer. In yet another embodiment, the composition is a tablet.
In yet another


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
13
embodiment, the composition is a lozenge. In yet another embodiment, the
composition is a
chewing gum. In yet another embodiment, the composition is a lipstick.

Methods for the preparation of solid pharmaceutical compositions are also well
known in the art.
Tablets may thus be prepared by mixing the active ingredient with ordinary
adjuvants, fillers and
diluents and subsequently compressing the mixture in a convenient tabletting
machine. Examples
of adjuvants, fillers and diluents comprise microcrystalline cellulose, corn
starch, potato starch,
lactose, mannitol, sorbitol talcum, magnesium stearate, gelatine, lactose,
gums, and the like. Any
other adjuvant or additive such as colorings, aroma, preservatives, etc. may
also be used provided
that they are compatible with the active ingredients.

In particular, the tablet formulations according to the invention may be
prepared by direct
compression of the compound of the invention with conventional adjuvants or
diluents.
Alternatively, a wet granulate or a melt granulate of the compound of the
invention, optionally in
admixture with conventional adjuvants or diluents may be used for compression
of tablets.

In a specific embodiment of the invention there is provided a pharmaceutical
composition
comprising a therapeutically effective amount of the compound of the
invention, or a
pharmaceutically acceptable acid addition salt thereof for administration via
the oral mucosa, in
particular buccally or sublingually.

Manufacturing processes for buccal and sublingual disintegrating tablets are
known in the art and
include, but are not limited to, conventional tableting techniques, freeze-
dried technology, and
floss-based tableting technology.
Conventional Tableting Techniques
Conventional tablet processing features conventional tablet characteristics
for ease of handling,
packaging, and fast disintegration (Ghosh and Pfister, Drug Delivery to the
Oral Cavity:
Molecule to Market, 2005, New York, CRC Press). The technology is based on a
combination of
physically modified polysaccharides that have water dissolution
characteristics that facilitate fast
disintegration and high compressibility. The result is a fast-disintegrating
tablet that has adequate
hardness for packaging in bottles and easy handling.

In certain embodiments, the manufacturing process involves granulating low-
moldable sugars
(e.g., mannitol, lactose, glucose, sucrose, and erythritol) that show quick
dissolution


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
14
characteristics with high-moldable sugars (e.g., maltose, sorbitol, trehalose,
and maltitol). The
result is a mixture of excipients that have fast-dissolving and highly
moldable characteristics
(Hamilton et at., Drug Deliv. Technol. 2005, 5, 34-37). The compound of the
invention can be
added, along with other standard tableting excipients, during the granulation
or blending
processes. The tablets are manufactured at a low compression force followed by
an optional
humidity conditioning treatment to increase tablet hardness (Parakh et at.,
Pharm. Tech. 2003,
27, 92-100).

In other embodiments, a compressed buccal or sublingual tablet comprising the
compound of the
invention is based on a conventional tableting process involving the direct
compression of active
ingredients, effervescent excipients, and taste-masking agents (see U.S.
5,223,614). The tablet
quickly disintegrates because effervescent carbon dioxide is produced upon
contact with
moisture. The effervescent excipient (known as effervescence couple) is
prepared by coating the
organic acid crystals using a stoichiometrically lesser amount of base
material. The particle size
of the organic acid crystals is carefully chosen to be larger than the base
excipient to ensure
uniform coating of the base excipient onto the acid crystals. The coating
process is initiated by
the addition of a reaction initiator, which is purified water in this case.
The reaction is allowed to
proceed only to the extent of completing the base coating on organic acid
crystals. The required
end-point for reaction termination is determined by measuring carbon dioxide
evolution. Then,
the excipient is mixed with the active ingredient or active microparticles and
with other standard
tableting excipients and then compressed into tablets.

In still other embodiments, the buccal or sublingual tablets are made by
combining non-
compressible fillers with a taste-masking excipient and active ingredient into
a dry blend. The
blend is compressed into tablets using a conventional rotary tablet press.
Tablets made with this
process have higher mechanical strength and are sufficiently robust to be
packaged in blister
packs or bottles (Aurora et at., Drug Deliv. Technol. 2005, 5:50-54). In other
embodiments, the
method further incorporates taste-masking sweeteners and flavoring agents such
as mint, cherry,
and orange. In certain embodiments, the compound of the invention tablets made
with this
process should disintegrate in the mouth in 5-45 seconds and can be formulated
to be
bioequivalent to intramuscular or subcutaneous dosage forms containing the
compound of the
invention.


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
Freeze-Dried Buccal or Sublingual Tablets
The freeze-drying process involves the removal of water (by sublimation upon
freeze drying)
from the liquid mixture of the compound of the invention matrix former, and
other excipients
filled into preformed blister pockets. The formed matrix structure is very
porous in nature and
5 rapidly dissolves or disintegrates upon contact with saliva (Sastry et at.,
Drug Delivery to the
Oral Cavity: Molecule to Market, 2005, New York, CRC Press, pp. 311-316).

Common matrix-forming agents include gelatins, dextrans, or alginates which
form glassy
amorphous mixtures for providing structural strength; saccharides such as
mannitol or sorbitol for
10 imparting crystallinity and hardness; and water, which functions as a
manufacturing process
medium during the freeze-drying step to induce the porous structure upon
sublimation. In
addition, the matrix may contain taste-masking agents such as sweeteners,
flavorants, pH-
adjusting agents such as citric acid, and preservatives to ensure the aqueous
stability of the
suspended drug in media before sublimation.
In this embodiment, freeze-dried buccal or sublingual Oral Disintegrating
Tablets (herein referred
to as ODTs) comprising the compound of the invention can be manufactured and
packaged in
polyvinyl chloride or polyvinylidene chloride plastic packs, or they may be
packed into laminates
or aluminum multilaminate foil pouches to protect the product from external
moisture.
Other known methods for manufacturing buccal or sublingual ODTs include
lyophilization (e.g.,
Lyoc (Farmalyoc, now Cephalon, Franzer, PA) and QuickSolv (Janssen
Pharmaceutica, Beerse,
Belgium). Lyoc is a porous, solid wafer manufactured by lyophilizing an oil-in-
water emulsion
placed directly in a blister and subsequently sealed. The wafer can
accommodate high drug
dosing and disintegrates rapidly but has poor mechanical strength (see EP
0159237). QuickSolv
tablets are made with a similar technology that creates a porous solid matrix
by freezing an
aqueous dispersion or solution of the matrix formulation. The process works by
removing water
using an excess of alcohol (solvent extraction). In certain embodiments, the
manufacturing
methods which utilize the lyophilization techniques, such as those related to
QuickSolv as
described above, could be of particular importance for producing buccal or
sublingual ODTs
comprising the compound of the invention. This is especially so in light of
the data provided
herein which shows the potential negative effect that highly water soluble
excipients can have in
the absorption of the compound of the invention in vivo. Thus, a buccal or
sublingual ODT
comprising the compound of the invention manufactured by such a lyophilization
technique


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
16
could provide increased in vivo absorption due of the removal of water soluble
excipients
occurring during the water removal step as described above.

Floss-Based Buccal or Sublingual Tablets
In other embodiments, floss-based tablet technology (e.g., FlashDose, Biovail,
Mississauga, ON,
Canada) can be used to produce fast-dissolving buccal or sublingual tablets
comprising the
compound of the invention using a floss known as the shearform matrix. This
floss is commonly
composed of saccharides such as sucrose, dextrose, lactose, and fructose. The
saccharides are
converted into floss by the simultaneous action of flash-melting and
centrifugal force in a heat-
processing machine similar to that used to make cotton candy. See U.S. Patents
5,587,172,
5,622,717, 5,567,439, 5,871,781, 5,654,003, and 5,622,716. The fibers produced
are usually
amorphous in nature and are partially re-crystallized, which results in a free-
flowing floss. The
floss can be mixed with the compound of the invention and pharmaceutically
acceptable
excipients followed by compression into a tablet that has fast- dissolving
characteristics.
Sublingual Tablets
Additional techniques can also be used to formulate the rapidly disintegrating
or dissolving
buccal or sublingual tablets of the present invention (Sastry et al., Pharm.
Sci. Tech. Today 2000,
3: 138-145; Chang et al., Pharmaceutical Technology 2000, 24: 52-58; Sharma et
at.,
Pharmaceutical Technology North America 2003, 10-15; Allen, International
Journal of
Pharmaceutical Technology 2003, 7, 449-450; Dobetti, Pharmaceutical Technology
Europe
2000, 12: 32-42; and Verma and Garg, Pharmaceutical Technology On-Line 2001,
25, 1-14).
Direct compression, one of these techniques, requires the incorporation of a
super disintegrant
into the formulation, or the use of highly water soluble excipients to achieve
fast tablet
disintegration or dissolution. Direct compression does not require the use of
moisture or heat
during tablet formation process, so it is very useful for the formulation and
compression of tablets
containing moisture-labile and heat-labile medications. However, the direct
compression method
is very sensitive to changes in the types and proportions of excipients, and
in the compression
force (CF), when used to achieve tablets of suitable hardness without
compromising the rapid
disintegration capabilities. As will be appreciated by one of skill in the
art, in order for tablets
administered sublingually to release the dose of medication for maximum rate
and extent of
absorption, the tablet must disintegrate almost instantaneously following
insertion into the
sublingual cavity. Precise selection and evaluation of the type and proportion
of excipients used


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
17
to formulate the tablet control the extent of hardness and rate of
disintegration. Compression
force (CF) can also be adjusted to result in tablets that have lower hardness
(H) and disintegrate
more quickly. Unique packaging methods such as strip packaging may be required
to compensate
for the problem of extreme friability of rapidly disintegrating, direct
compression tablets.
Watenabe et at. (Watanabe et at., Biol. Pharm. Bull. 1995, 18: 1308-1310;
Ishikawa et at., Chem.
Pharm. Bull. 2001, 49: 134-139) and Bi et al (Bi et at., Chem. Pharm. Bull.
1996, 44: 2121-2127;
Bi et at., Drug Dev. Lnd. Pharm. 1999, 25: 571-581) were the first to evaluate
the ideal excipient
proportions and other related parameters required to formulate durable fast
disintegrating tablets
using a super disintegrant. They studied the effect of a wide range of
microcrystalline cellulose:
low-substituted hydroxypropyl cellulose (MCC:L HPC) ratios on the tablet
characteristics.

In a further aspect the invention provides the use of said composition for the
preparation of a
medicament for the treatment of neurodegenerative disorders such as
Parkinson's disease and
Huntington's disease.

In a further aspect the invention provides the use of the pharmaceutical
composition for the
preparation of a medicament for the treatment of psychoses, impotence, renal
failure, heart failure
or hypertension.
In another aspect the invention provides the use of the pharmaceutical
composition for the
manufacture of a medicament for the treatment of cognitive impairment in a
mammal.

In a still further aspect the invention provides the use of the pharmaceutical
composition for the
manufacture of a medicament for the treatment of restless legs syndrome (RLS)
or periodic limb
movement disorder (PLMD).

In a still further aspect the invention provides the use of the pharmaceutical
composition for the
manufacture of a medicament for the treatment of erectile dysfunction.
In a different aspect the invention provides the use of the pharmaceutical
composition for the
manufacture of a medicament for the treatment of movement disorders, poverty
of movement,
dyskinetic disorders, gait disorders or intention tremor in a mammal.


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
18
In a further aspect the invention provides the use of the pharmaceutical
composition for the
treatment of neurodegenerative disorders such as Parkinson's disease and
Huntington's disease.

In a further aspect the invention provides the use of the pharmaceutical
composition for the
treatment of psychoses, impotence, renal failure, heart failure or
hypertension.

In another aspect the invention provides the use of the pharmaceutical
composition for the
treatment of cognitive impairment in a mammal.

In a still further aspect the invention provides the use of the pharmaceutical
composition for the
treatment of restless legs syndrome (RLS) or periodic limb movement disorder
(PLMD).

In a different aspect the invention provides the use of the pharmaceutical
composition for the
treatment of movement disorders, poverty of movement, dyskinetic disorders,
gait disorders or
intention tremor in a mammal.

In separate aspects the invention provides the use of the pharmaceutical
composition for the
manufacture of medicaments, which are intended for administration via the oral
mucosa.

The invention also provides a method of treating a mammal suffering from a
neurodegenerative
disorder such as Parkinson's disease and Huntington's disease comprising
administering to the
mammal a therapeutically effective amount of the pharmaceutical composition.

In another aspect the invention also provides a method of treating a mammal
suffering from
psychoses, impotence, renal failure, heart failure or hypertension, comprising
administering to the
mammal a therapeutically effective amount of the pharmaceutical composition.

In a further aspect the invention provides a method of treating a mammal
suffering from a
cognitive impairment, comprising administering to the mammal an effective
amount of the
pharmaceutical composition.

The invention also relates to a method of treating a mammal suffering from
restless legs
syndrome (RLS) or periodic limb movement disorder (PLMD), comprising
administering to the
mammal a therapeutically effective amount of the compound of the invention, or
a
pharmaceutically acceptable addition salt thereof


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
19
The invention also relates in a separate aspect to a method of treating a
mammal suffering from
movement disorders, poverty of movement, dyskinetic disorders, gait disorders
or intention
tremor comprising administering to the mammal of the pharmaceutical
composition.
The therapeutically effective amount of the compound of the invention,
calculated as the daily
dose of the compound of the invention above as the free base, is suitably
between 0.001 and 12.5
mg/day, more suitable between 0.005 and 10.0 mg/day, e.g. preferably between
0.01 and 5.0
mg/day. In a specific embodiment the daily dose of the compound of the
invention is between 0.1
and 1.0 mg/day.

In another embodiment the daily dose of the compound of the invention is less
than about 0.1
mg/day. In a separate embodiment the daily dose of the compound of the
invention is about 0.01
mg/day. In a further embodiment the invention provides a formulation
comprising from 0.0001
mg to 12.5 mg of the compound of the invention for delivery via the oral
mucosa. In a further
embodiment the invention provides a formulation comprising from 0.0001 mg to
0.01 mg of the
compound of the invention for delivery via the oral mucosa. In a further
embodiment the
invention provides a formulation comprising from 0.001 mg to 0.10 mg of the
compound of the
invention for delivery via the oral mucosa. In a further embodiment the
invention provides a
formulation comprising from 0.01 mg to 1.0 mg of the compound of the invention
for delivery
via the oral mucosa.

In yet other embodiments, the invention described herein provides
pharmaceutical tablets for
buccal or sublingual administration comprising the compound of the invention
wherein the
administration of the pharmaceutical tablets provides a pharmacokinetic
profile substantially
equivalent to the pharmacokinetic profile of traditional injectable dosage
forms comprising the
compound of the invention administered either subcutaneously or
intramuscularly. In certain
embodiments, the pharmaceutical tablets for buccal or sublingual
administration described herein
can provide a pharmacokinetic profile substantially equivalent to the
pharmacokinetic profile of
traditional injectable dosage forms comprising the compound of the invention
administered either
subcutaneously or intramuscularly, wherein the pharmacokinetic profile
consists of one or more
of the pharmacokinetic parameters selected from the group consisting of.
C,,,a.X, T,,,aX, AUC(tast),
and AUC(o~).


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
Ultimately, the exact dose of the compound of the invention and the particular
formulation to be
administered depend on a number of factors, e.g., the condition to be treated,
the desired duration
of the treatment and the rate of release of the active agent. For example, the
amount of the active
agent required and the release rate thereof may be determined on the basis of
known in vitro or in
5 vivo techniques, determining how long a particular active agent
concentration in the blood plasma
remains at an acceptable level for a therapeutic effect.

B. Intranasal Administration
The term "intranasal delivery" as used herein means a method for drug
absorption through and
10 within the nasal mucosa.

Carriers" or "vehicles" as used herein refer to carrier materials suitable for
intranasal drug
administration, and include any such materials known in the art, e.g., any
liquid, gel, solvent,
liquid diluent, solubilizer, or the like, which is non toxic and which does
not interact with other
15 components of the composition in a deleterious manner. Examples of suitable
vehicles for use
herein include water, alcohols such as isopropyl alcohol and isobutyl alcohol,
polyalcohol such as
glycerol, and glycols such as propylene glycol, and esters of such polyols,
(e.g., mono-, di-, or tri-
glycerides).

20 Intranasal Compositions
Relative to an oral dosage form such as a tablet or capsule, intranasal
delivery provides for rapid
absorption, faster onset of therapeutic action and avoidance of gut wall or
liver first pass
metabolism. For patients who have difficulty in swallowing tablets, capsules
or other solids or
those who have intestinal failure, the intranasal delivery route may be
preferred.
The compositions for nasal administration include the compound of the
invention, or a
pharmaceutically acceptable salt thereof, and optionally can also include
other ingredients
including, but not limited to, carriers and excipients, such as absorption-
promoting agents which
promote nasal absorption of the active ingredient after nasal administration.
Other optional
excipients include diluents, binders, lubricants, glidants, disintegrants,
desensitizing agents,
emulsifiers, mucosal adhesives, solubilizers, suspension agents, viscosity
modifiers, ionic tonicity
agents, buffers, carriers, flavors and mixtures thereof.


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
21
The amount of drug absorbed depends on many factors. These factors include the
drug
concentration, the drug delivery vehicle, mucosal contact time, the venous
drainage of the
mucosal tissues, the degree that the drug is ionized at the pH of the
absorption site, the size of the
drug molecule, and its relative lipid solubility. Those of skill in the art
can readily prepare an
appropriate intranasal composition, which delivers an appropriate amount of
the active agent,
taking these factors into consideration.

Absorption Promoting Agents
The transport of the active ingredient across normal nasal mucosa can be
enhanced by optionally
combining it with an absorption promoting agent, such as those disclosed in
U.S. Patent Nos.
5,629,011, 5,023,252, 6,200,591, 6,369,058, 6,380,175, and International
Publication Number
WO 01/60325. Examples of these absorption promoting agents include, but are
not limited to,
cationic polymers, surface active agents, chelating agents, mucolytic agents,
cyclodextrin,
polymeric hydrogels, combinations thereof, and any other similar absorption
promoting agents
known to those of skill in the art. Representative absorption promoting
excipients include
phospholipids, such as phosphatidylglycerol or phosphatidylcholine,
lysophosphatidyl
derivatives, such as lysophosphatidylethanolamine, lysophosphatidylcho line,
lysophosphatidylglycerol, lysophosphatidylserine, or lysophosphatidic acid,
polyols, such as
glycerol or propylene glycol, fatty acid esters thereof such as glycerides,
amino acids, and esters
thereof, and cyclodextrins. Gelling excipients or viscosity-increasing
excipients can also be used.
MucoadhesiveBioadhesive Polymers
The transport of the active ingredient across normal mucosal surfaces can also
be enhanced by
increasing the time in which the formulations adhere to the mucosal surfaces.
Mucoadhesive/bioadhesive polymers, for example, those which form hydrogels,
exhibit muco-
adhesion and controlled drug release properties and can be included in the
intranasal
compositions described herein. Examples of such formulations are disclosed in
U.S. Patent Nos.
6,068,852 and 5,814,329; and International Publication Number W099/58110.
Representative
bioadhesive or hydrogel-forming polymers capable of binding to the nasal
mucosa are well
known to those of skill in the art, and include polycarbophil, polylysine,
methylcellulose, sodium
carboxymethylcellulose, hydroxypropyl-methylcellulose, hydroxyethyl cellulose,
pectin,
Carbopol 934P, polyethylene oxide 600K, Pluronic F127, polyisobutylene (PIB),
polyisoprene


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
22
(PIP), polyvinyl pyrrolidone (PVP), polyvinyl alcohol (PVA), xanthum gum, guar
gum, and
locust bean gum.

Other nasal delivery compositions are chitosan-based and are suitable to
increase the residence
time of the active ingredient on mucosal surfaces, which results in increasing
its bioavailability.
Examples of these nasal delivery compositions are disclosed in U.S. Patent
Nos. 6,465,626,
6,432,440, 6,391 ,318, and 5,840,341; European Patent Numbers EP0993483 and
EP1051190;
and International Publication Numbers WO 96/05810, WO 96/03142, and WO
93/15737.
Additionally, the present invention can be formulated with powder microsphere
and
mucoadhesive compositions as disclosed in European Patent Numbers EP1025859
and
EP 1108423, which are incorporated herein by reference with regard to such
composition.

Finally, thiolated polymeric excipients that form covalent bonds with the
cysteine-rich
subdomains of the mucus membrane can also provide mucoadhesion, which prolongs
the contact
time between the active ingredient and the membrane. Such excipients are
disclosed in
International Publication Number WO 03/020771.

Antioxidants
The buccal compositions can also include one or more antioxidants.
Representative antioxidants
include quaternary ammonium salts such as lauralkonium chloride, benzalkonium
chloride,
benzododecinium chloride, cetyl pyridium chloride, cetrimide, domiphen
bromide; alcohols such
as benzyl alcohol, chlorobutanol, o-cresol, phenyl ethyl alcohol; organic
acids or salts thereof
such as ascorbic acid, benzoic acid, sodium benzoate, sodium ascorbate,
potassium sorbate,
parabens; or complex forming agents such as ethylenediaminetetraacetic acid
(EDTA).
Other Excipients
The carriers and excipients include ion-exchange microspheres which carry
suitable anionic
groups such as carboxylic acid residues, carboxymethyl groups, sulphopropyl
groups and
methylsulphonate groups. Ion-exchange resins, such as cation exchangers, can
also be used.
Chitosan, which is partially deacetylated chitin, or poly-N-acetyl-D-
glucosamine, or a
pharmaceutically acceptable salt thereof such as hydrochloride, lactate,
glutamate, maleate,
acetate, formate, propionate, maleate, malonate, adipate, or succinate.
Suitable other ingredients
for use as non-ion-exchange microspheres include starch, gelatin, collagen and
albumin.


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
23
The composition can also include an appropriate acid selected from the group
consisting of
hydrochloric acid, lactic acid, glutamic acid, maleic acid, acetic acid,
formic acid, propionic acid,
malic acid, malonic acid, adipic acid, and succinic acid. Other ingredients
such as diluents are
cellulose, microcrystalline cellulose, hydroxypropyl cellulose, starch,
hydroxypropylmethyl
cellulose, and the like.

Excipients to adjust the tonicity of the composition may be added such as
sodium chloride,
glucose, dextrose, mannitol, sorbitol, lactose, and the like. Acidic or basic
buffers can also be
added to the intranasal composition to control the pH.
Incorporation of the Active Agent into the Compositions
In addition to using absorption enhancing agents, which increase the transport
of the active agents
through the mucosa, and bioadhesive materials, which prolong the contact time
of the active
agent along the mucosa, the administration of the active agent can be
controlled by using
controlled release formulations, which can provide rapid or sustained release,
or both, depending
on the formulations.

There are numerous particulate drug delivery vehicles known to those of skill
in the art which can
include the active ingredients, and deliver them in a controlled manner.
Examples include
particulate polymeric drug delivery vehicles, for example, biodegradable
polymers, and particles
formed of non-polymeric components. These particulate drug delivery vehicles
can be in the
form of powders, microparticles, nanoparticles, microcapsules, liposomes, and
the like.
Typically, if the active agent is in particulate form without added
components, its release rate
depends on the release of the active agent itself. Typically, the rate of
absorption is enhanced by
presenting the drug in a micronized form, wherein particles are below 20
microns in diameter. In
contrast, if the active agent is in particulate form as a blend of the active
agent and a polymer, the
release of the active agent is controlled, at least in part, by the removal of
the polymer, typically
by dissolution, biodegradation, or diffusion from the polymer matrix.

The compositions can provide an initial rapid release of the active ingredient
followed by a
sustained release of the active ingredient. U.S. Patent No. 5,629,011 provides
examples of this
type of formulation and is incorporated herein by reference with regard to
such formulations.


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
24
There are numerous compositions that utilize intranasal delivery and related
methods thereof
Moreover, there are numerous methods and related delivery vehicles that
provide for intranasal
delivery of various pharmaceutical compositions. For example, intranasal
compositions that
employ current marketed nicotine replacement therapies (See, N. J. Benowitz,
Drugs, 45: 157-
170 (1993) are also suitable for administering the compounds described herein.
Nasal Insufflator Devices
The intranasal compositions can be administered by any appropriate method
according to their
form. A composition including microspheres or a powder can be administered
using a nasal
insufflator device. Examples of these devices are well known to those of skill
in the art, and
include commercial powder systems such as Fisons Lomudal System. An
insufflator produces a
finely divided cloud of the dry powder or microspheres. The insufflator is
preferably provided
with a mechanism to ensure administration of a substantially fixed amount of
the composition.
The powder or microspheres can be used directly with an insufflator, which is
provided with a
bottle or container for the powder or microspheres. Alternatively, the powder
or microspheres can
be filled into a capsule such as a gelatin capsule, or other single dose
device adapted for nasal
administration. The insufflator preferably has a mechanism to break open the
capsule or other
device.

Further, the composition can provide an initial rapid release of the active
ingredient followed by a
sustained release of the active ingredient, for example, by providing more
than one type of
microsphere or powder.

Use of Metered Sprays
Intranasal delivery can also be accomplished by including the active
ingredient in a solution or
dispersion in an aqueous medium which can be administered as a spray.
Appropriate devices for
administering such a spray include metered dose aerosol valves and metered
dose pumps,
optionally using gas or liquid propellants.

Representative devices of this type are disclosed in the following patents,
patent applications, and
publications: WO 03/026559, WO 02/011800, WO 00/51672, WO 02/068029, WO
02/068030,
WO 02/068031, WO 02/068032, WO 03/000310, WO 03/020350, WO 03/082393, WO
03/084591, WO 03/090812, WO 00/41755, and the pharmaceutical literature (See,
Bell, A.
Intranasal Delivery Devices, in Drug Delivery Devices Fundamentals and
Applications, Tyle P.


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
(ed), Dekker, New York, 1988); and Remington's Pharmaceutical Sciences, Mack
Publishing
Co., 1975.

Other Modes of Intranasal Delivery
5 In addition to the foregoing, the compounds and intranasal compositions
including the
compounds can also be administered in the form of nose-drops, sprays,
irrigations, and douches,
as is known in the art. Nose drops are typically administered by inserting
drops while lying on a
bed, with the patient on his or her back, especially with the head lying over
the side of the bed.
This approach helps the drops get farther back.
Nasal irrigation involves regularly flooding the nasal cavity with warm salty
water, which
includes one or more compounds as described herein, or their pharmaceutically
acceptable salts.
Nasal douches are typically used by filling a nasal douche with a salt
solution including one or
more compounds as described herein, or their pharmaceutically acceptable
salts, inserting the
nozzle from the douche into one nostril, opening one's mouth to breathe, and
causing the solution
to flow into one nostril, rinse round the septum and turbinates, and discharge
from the other
nostril.

As mentioned previously, the present invention provides pharmaceutical
compositions for
intranasal administration of (4aR,1OaR)-1-n-propyl-1,2,3,4,4a,5,10,1Oa-
octahydro-
benzo[g]quinoline-6,7-diol and related compounds, which may be delivered to
the systemic
circulation via delivery across the nasal mucosa.

In one embodiment, the composition is further comprising an absorption agent.
In one
embodiment, the composition is further comprising one or more adhesive,
binder, lubricant,
glidant, disintegrant or mixture thereof.

The compound of the invention as a pharmaceutical composition for intranasal
adminstration
may be administered in any suitable way in the nasal cavity, and the compound
may be presented
in any suitable dosage form for such administration, e.g. in form of simple
solutions or
dispersions, simple tablets, matrix tablets, capsules, powders, syrups,
dissolvable films, patches,
lipophilic gels. In one embodiment, the compound of the invention is
administered in the form of
a solid pharmaceutical entity, suitably as a tablet or a capsule. In another
particular embodiment,
the compound of the invention is administered in the form of a dissolvable
film.


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
26
In the case of intranasal administration of the compound of the invention,
conventional dosage
forms may not be able to assure therapeutic drug levels in because of
physiological removal
mechanism of the oral cavity (washing effect of saliva and mechanical stress),
which remove the
drug formulation away from the nasal mucosa, resulting in too short exposure
time and
unpredictable absorption. To obtain the desired therapeutic action it may
therefore be necessary
to prolong and improve the contact between the compound of the invention and
the nasal
mucosa. To fulfill the therapeutic requirement, formulations designed for
intranasal
administration may therefore contain mucoadhesive agents to maintain an
intimate and prolonged
contact of the formulation with the absorption site; penetration enhancers, to
improve drug
permeation across the mucosa; and enzyme inhibitors to eventually protect the
drug from
degradation by means of nasal mucosal enzymes.

In a specific embodiment of the invention there is provided a pharmaceutical
composition
comprising a therapeutically effective amount of compound of the invention or
a
pharmaceutically acceptable acid addition salt thereof for administration via
the nasal mucosa.

In a further aspect the invention provides the use of said composition for the
preparation of a
medicament for the treatment of neurodegenerative disorders such as
Parkinson's disease and
Huntington's disease.

In a further aspect the invention provides the use of the pharmaceutical
composition for the
preparation of a medicament for the treatment of psychoses, impotence, renal
failure, heart failure
or hypertension.
In another aspect the invention provides the use of the pharmaceutical
composition for the
manufacture of a medicament for the treatment of cognitive impairment in a
mammal.

In a still further aspect the invention provides the use of the pharmaceutical
composition for the
manufacture of a medicament for the treatment of restless legs syndrome (RLS)
or periodic limb
movement disorder (PLMD).

In a still further aspect the invention provides the use of the pharmaceutical
composition for the
manufacture of a medicament for the treatment of erectile dysfunction.


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
27
In a different aspect the invention provides the use of the pharmaceutical
composition for the
manufacture of a medicament for the treatment of movement disorders, poverty
of movement,
dyskinetic disorders, gait disorders or intention tremor in a mammal.

In a further aspect the invention provides the use of the pharmaceutical
composition for the
treatment of neurodegenerative disorders such as Parkinson's disease and
Huntington's disease.

In a further aspect the invention provides the use of the pharmaceutical
composition for the
treatment of psychoses, impotence, renal failure, heart failure or
hypertension.
In another aspect the invention provides the use of the pharmaceutical
composition for the
treatment of cognitive impairment in a mammal.

In a still further aspect the invention provides the use of the pharmaceutical
composition for the
treatment of restless legs syndrome (RLS) or periodic limb movement disorder
(PLMD).

In a different aspect the invention provides the use of the pharmaceutical
composition for the
treatment of movement disorders, poverty of movement, dyskinetic disorders,
gait disorders or
intention tremor in a mammal.
In separate aspects the invention provides the use of the pharmaceutical
composition for the
manufacture of medicaments, which are intended for administration via the oral
mucosa.

The invention also provides a method of treating a mammal suffering from a
neurodegenerative
disorder such as Parkinson's disease and Huntington's disease comprising
administering to the
mammal a therapeutically effective amount of the pharmaceutical composition.

In another aspect the invention also provides a method of treating a mammal
suffering from
psychoses, impotence, renal failure, heart failure or hypertension, comprising
administering to the
mammal a therapeutically effective amount of the pharmaceutical composition.

In a further aspect the invention provides a method of treating a mammal
suffering from a
cognitive impairment, comprising administering to the mammal an effective
amount of the
pharmaceutical composition.


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
28
The invention also relates to a method of treating a mammal suffering from
restless legs
syndrome (RLS) or periodic limb movement disorder (PLMD), comprising
administering to the
mammal a therapeutically effective amount of compound of the invention, or a
pharmaceutically
acceptable addition salt thereof
The invention also relates in a separate aspect to a method of treating a
mammal suffering from
movement disorders, poverty of movement, dyskinetic disorders, gait disorders
or intention
tremor comprising administering to the mammal of the pharmaceutical
composition.

The therapeutically effective amount of the compound of the invention,
calculated as the daily
dose of the compound of the invention above as the free base, is suitably
between 0.001 and 12.5
mg/day, more suitable between 0.005 and 10.0 mg/day, e.g. preferably between
0.01 and 5.0
mg/day. In a specific embodiment the daily dose of the compound of the
invention is between 0.1
and 1.0 mg/day.
In another embodiment the daily dose of the compound of the invention is less
than about 0.1
mg/day. In a separate embodiment the daily dose of the compound of the
invention is about 0.01
mg/day. In a further embodiment the invention provides a formulation
comprising from 0.0001
mg to 12.5 mg of the compound of the invention for delivery via the nasal
mucosa. In a further
embodiment the invention provides a formulation comprising from 0.0001 mg to
0.01 mg of the
compound of the invention for delivery via the nasal mucosa. In a further
embodiment the
invention provides a formulation comprising from 0.001 mg to 0.10 mg of the
compound of the
invention for delivery via the nasal mucosa. In a further embodiment the
invention provides a
formulation comprising from 0.01 mg to 1.0 mg of the compound of the invention
for delivery
via the nasal mucosa.

C. Transdermal Administration
By "transdermal delivery", applicants intend to include both transdermal and
percutaneous
administration, i.e., delivery by passage of an active ingredient through the
skin and into the
bloodstream.

"Carriers" or "vehicles" as used herein refer to carrier materials suitable
for transdermal drug
administration, and include any such materials known in the art, e.g., any
liquid, gel, solvent,
liquid diluent, solubilizer, or the like, which is non toxic and which does
not interact with other


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
29
components of the composition in a deleterious manner. Examples of suitable
vehicles for use
herein include water, alcohols such as isopropyl alcohol and isobutyl alcohol,
polyalcohols such
as glycerol, and glycols such as propylene glycol, and esters of such polyols,
(e.g., mono-, di-, or
tri-glycerides).
"Penetration enhancement" or "permeation enhancement" as used herein relates
to an increase in
the permeability of skin to a pharmacologically active agent, namely, so as to
increase the rate at
which the active ingredient permeates through the skin (i.e., flux) and enters
the bloodstream or
the local site of action. The enhanced permeation effected by using these
enhancers can be
observed by measuring the rate of diffusion (or flux) of active ingredient
through animal or
human skin or a suitable polymeric membrane using a diffusion cell apparatus
as described in the
examples herein.

Permeation enhancers are described, for example, in U.S. Patent Nos.
5,785,991; 4,764,381;
4,956,171; 4,863,970; 5,453,279; 4,883,660; 5,719,197, and in the literature
"Pharmaceutical
Skin Penetration Enhancement", J. Hadgraft, Marcel Dekker, Inc. 1993;
"Percutaneous
Absorption", R. Bronaugh, H. Maibach, Marcel Dekker, Inc. (1989), B. W. Barry,
"Penetration
Enhancers in Skin Permeation", Proceedings of the 13th international Symposium
on Controlled
Release of Bioactive Materials, ed. by Chaudry & Thies, Controlled Release
Society,
Lincolnshire, III., pp. 136-137 (1986), and Cooper & Berner, "Penetration
Enhancers", in The
Transdermal Delivery of Ingredients, Vol. Il ed. by Kydonieus and Berner, CRC
Press, Boca
Raton, Fla. pp. 57-62 (1986).

The permeation enhancers should both enhance the permeability of the stratum
corneum, and be
non-toxic, non-irritant and non-sensitizing on repeated exposure.
Representative permeation
enhancers include, for example, sucrose monococoate, glycerol monooleate,
sucrose
monolaurate, glycerol monolaureate, diethylene glycol monoalkyl ethers such as
diethylene
glycol monoethyl or monomethyl ether (Transcutol P), ester components such as
propylene
glycol monolaurate, methyl laurate, and lauryl acetate, monoglycerides such as
glycerol
monolaurate, fatty alcohols such as lauryl alcohol, and 2-ethyl-1,3 hexanediol
alone or in
combination with oleic acid.


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
Gelling Agents
Gelling agents, such as carbomer, carboxyethylene or polyacrylic acid such as
Carbopol 980 or
940 NF, 981 or 941 NF, 1382 or 1342 NF, 5984 or 934 NF, ETD 2020, 2050, 934P
NF, 971 P
NF, 974P NF, Noveon AA-1 USP, etc; cellulose derivatives such as
ethylcellulose,
5 hydroxypropylmethylcellulose (HPMC), ethylhydroxyethylcellulose (EHEC),
carboxymethylcellulose (CMC), hydroxypropylcellulose (HPC) (Klucel , different
grades),
hydroxyethylcellulose (HEC) (Natrosol grades), HPMCP 55, Methocel grades,
etc; natural
gums such as arabic, xanthan, guar gums, alginates, etc; polyvinylpyrrolidone
derivatives such as
Kollidon grades; polyoxyethylene polyoxypropylene copolymers such as Lutrol
F grades 68,
10 127, etc; others like chitosan, polyvinyl alcohols, pectins, veegun grades,
and the like, can also be
present. Those of the skill in the art know of other gelling agents or
viscosants suitable for use in
the present invention. Representative gelling agents include, but are not
limited to, Carbopol
980 NF, Lutrol F 127, Lutrol F 68 and Noveon AA-1 USP. The gelling agent is
present
from about 0.2 to about 30.0% w/w, depending on the type of polymer.
Antioxidants
The transdermal compositions can also include one or more antioxidants.
Representative
antioxidants include quaternary ammonium salts such as lauralkonium chloride,
benzalkonium
chloride, benzododecinium chloride, cetyl pyridium chloride, cetrimide,
domiphen bromide;
alcohols such as benzyl alcohol, chlorobutanol, o-cresol, phenylethyl alcohol;
organic acids or
salts thereof such as ascorbic acid, benzoic acid, sodium ascorbate, sodium
benzoate, potassium
sorbate, parabens; or complex forming agents such as
ethylenediaminetetraacetic acid (EDTA).
Representative antioxidants include butylhydroxytoluene, butylhydroxyanisole,
ethylenediaminetetraacetic acid and its sodium salts, D,L-alpha tocoferol.
Other Components
Other components may include diluents such as cellulose, microcrystalline
cellulose,
hydroxypropyl cellulose, starch, hydroxypropylmethyl cellulose and the like.
Excipients can be
added to adjust the tonicity of the composition, such as sodium chloride,
glucose, dextrose,
mannitol, sorbitol, lactose and the like. Acidic or basic buffers can also be
added to control the
pH. Co-solvents or solubilizers such as glycerol, polyethylene glycols,
polyethylene glycols
derivatives, polyethylene glycol 660 hydroxystearate (Solutol HS15 from BASF),
butylene
glycol, hexylene glycol, and the like, can also be added.


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
31
Transdermal Compositions
The compositions for transdermal administration include a compound of the
invention including
fatty acid salts, and optionally can also include other ingredients including,
but not limited to,
carriers and excipients, such as permeation enhancers which promote
transdermal absorption of
the active ingredient after transdermal administration.

The amount of active ingredient absorbed depends on many factors. These
factors include the
active ingredient concentration, the active ingredient delivery vehicle, the
skin contact time, the
area of the skin dosed, the ratio of the ionized and unionized forms of the
active ingredient at the
pH of the absorption site, the molecular size of the active ingredient
molecule, and the active
ingredient's relative lipid solubility.

Transdermal Devices
The transdermal device for delivering the active ingredients described herein
can be of any type
known in the art, including the monolithic, matrix, membrane, and other types
typically useful for
administering active ingredients by the transdermal route. Such devices are
disclosed in U.S. Pat.
Nos. 3,996,934; 3,797,494; 3,742,951; 3,598,122; 3,598,123; 3,731,683;
3,734,097; 4,336,243;
4,379,454; 4,460,372; 4,486,193; 4,666,441; 4,615,699; 4,681,584; and
4,558,580 among others.

These devices tend to be flexible, adhere well to the skin, and have a
polymeric backing
(covering) that is impermeable to the active ingredient to be delivered, so
that the active
ingredient is administered uni-directionally through the skin. The active
ingredient, or
pharmaceutically acceptable salt thereof, is typically present in a solution
or dispersion, which
can be in the form of a gel, a solution, or a semi-solid, and which aids in
active ingredient
delivery through the stratum corneum of the epidermis and to the dermis for
absorption.

Membrane Devices
Membrane devices typically have four layers: (1) an impermeable backing, (2) a
reservoir layer,
(3) a membrane layer (which can be a dense polymer membrane or a microporous
membrane),
and (4) a contact adhesive layer which either covers the entire device surface
in a continuous or
discontinuous coating or surrounds the membrane layer. Examples of materials
that may be used
to act as an impermeable layer are high, medium, and low density polyethylene,
polypropylene,
polyvinylchloride, polyvinylidene chloride, polycarbonate, polyethylene
terepthalate, and
polymers laminated or coated with aluminum foil. Others are disclosed in the
standard


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
32
transdermal device patents mentioned herein. In certain embodiments in which
the reservoir layer
is fluid or is a polymer, the outer edge of the backing layer can overlay the
edge of the reservoir
layer and be sealed by adhesion or fusion to the diffusion membrane layer. In
such instances, the
reservoir layer need not have exposed surfaces.
The reservoir layer is underneath the impermeable backing and contains a
carrier liquid, typically
water and/or an alcohol, or polyol or ester thereof, and may or may not
contain the active
ingredients. The reservoir layer can include diluents, stabilizers, vehicles,
gelling agents, and the
like in addition to the carrier liquid and active ingredients.
The diffusion membrane layer of the laminate device can be made of a dense or
microporous
polymer film that has the requisite permeability to the active ingredient and
the carrier liquid.
Preferably, the membrane is impermeable to ingredients other than the active
ingredient and the
carrier liquid, although when buffering at the skin surface is desired, the
membrane should be
permeable to the buffer in the composition as well. Examples of polymer film
that may be used to
make the membrane layer are disclosed in U.S. Pat. Nos. 3,797,454 and
4,031,894. The preferred
materials are polyurethane, ethylene vinyl alcohol polymers, and
ethylene/vinyl acetate.
Monolithic Matrices
The second class of transdermal systems is represented by monolithic matrices.
Examples of such
monolithic devices are U.S. Pat. Nos. 4,291,014; 4,297,995; 4,390,520 and
4,340,043. Others are
known to those of ordinary skill in this art.

Monolithic and matrix type barrier transdermal devices typically include: (1)
Porous polymers or
open-cell foam polymers, such as polyvinyl chloride (PVC), polyurethanes,
polypropylenes, and
the like; (2) Highly swollen or plasticized polymers such as cellulose, HEMA
or MEMA or their
copolymers, hydroxypropyl methylcellulose (HPMC), hydroxyethyl methylcellulose
(HEMC),
and the like, polyvinyl alcohol (PVA)/ polyvinylpyrollidone (PVP), or other
hydrogels, or PVC,
polyurethane, ethylene / vinyl acetate, or their copolymers; (3) Gels of
liquids, typically including
water and/or hydroxyl-containing solvents such as ethanol, and often
containing gelling agents
such PVP, carboxymethylcellulose (CMC), hydroxypropylcellulose such as sold
under the
tradename Klucel , HPMC, alginates, kaolinate, bentonite, or montmorillonite,
other clay fillers,
stearates, silicon dioxide particles, and the like; (4) Nonwoven materials
made of textiles,
celluloses, polyurethanes, polyester, or other fiber; (5) Sponges, which can
be formed from


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
33
natural or foamed polymers; and (6) Adhesives, ideally dermatologically-
acceptable pressure
sensitive adhesives, for example, silicone adhesives or acrylic adhesives.

Polymeric Barrier Materials
Representative polymeric barrier materials include, but are not limited to:
Polycarbonates, such
as those formed by phosgenation of a dihydroxy aromatic such as bisphenol A,
including
materials are sold under the trade designation Lexan (the General Electric
Company);
Polyvinylchlorides, such as Geon 121 (B. G. Goodrich Chemical Company);
Polyamides
("nylons"), such as polyhexamethylene adipamide, including NOMEX (E. I.
DuPont de
Nemours & Co.).

Modacrylic copolymers, such as DYNEL , are formed of polyvinylchloride (60
percent) and
acrylonitrile (40 percent), styrene-acrylic acid copolymers, and the like.
Polysulfones, for
example, those containing diphenylene sulfone groups, for example, P-1700
(Union Carbide
Corporation). Halogenated polymers, for example, polyvinylidene fluoride, such
as Kynar
(Pennsalt Chemical Corporation), polyvinylfluoride, such as Tedlar (E. I.
DuPont de Nemours
& Co.), and polyfluorohalocarbons, such as Aclar (Allied Chemical
Corporation).
Polychlorethers, for example, Penton (Hercules Incorporated), and other
thermoplastic
polyethers. Acetal polymers, for example, polyformaldehydes, such as Delrin
(E. I. DuPont de
Nemours & Co.). Acrylic resins, for example, polyacrylonitrile, polymethyl
methacrylate
(PMMA), poly n-butyl methacrylate, and the like.

Other polymers such as polyurethanes, polyimides, polybenzimidazoles,
polyvinyl acetate,
aromatic and aliphatic, polyethers, cellulose esters, e.g., cellulose
triacetate; cellulose; colledion
(cellulose nitrate with 11% nitrogen); epoxy resins; olefins, e.g.,
polyethylene, polypropylene;
polyvinylidene chloride; porous rubber; cross linked poly(ethylene oxide);
cross-linked
polyvinylpyrrolidone; cross-linked polyvinyl alcohol); polyelectrolyte
structures formed of two
ionically associated polymers of the type as set forth in U.S. Pat. Nos.
3,549,016 and 3,546,141;
derivatives of polystyrene such as poly(sodium styrenesulfonate) and
poly(vinylbenzyltrimethyl-
ammonium chloride); poly(hydroxyethylmethacrylate); poly(isobutylvinyl ether),
and the like,
may also be used. A large number of copolymers which can be formed by reacting
various
proportions of monomers from the above list of polymers are also useful. If
the membrane or
other barrier does not have a sufficiently high flux, the thickness of the
membrane or barrier can


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
34
be reduced. However, the thickness should not be reduced to the point where it
is likely to tear, or
to a point where the amount of active ingredient which can be administered is
too low.

Adhesives
The transdermal drug delivery compositions typically include a contact
adhesive layer to adhere
the device to the skin. The active agent may, in some embodiments, reside in
the adhesive.
Adhesives include polyurethanes; acrylic or methacrylic resins such as
polymers of esters of
acrylic or methacrylic acid with alcohols such as n- butanol, n-pentanol,
isopentanol, 2-
methylbutanol, 1 -methylbutanol, 1-methylpentanol, 2-methylpentanol, 3-
methylpentanol, 2-
ethylbutanol, isooctanol, n-decanol, or n-dodecanol, alone or copolymerized
with ethylenically
unsaturated monomers such as acrylic acid, methacrylic acid, acrylamide,
methacrylamide, N-
alkoxymethyl acrylamides, N-alkoxymethyl methacrylamides, N-
tertbutylacrylamide, itaconic
acid, vinylacetate, N-branched alkyl maleamic acids wherein the alkyl group
has 10 to 24 carbon
atoms, glycol diacrylates, or mixtures of these; natural or synthetic rubbers
such as
styrenebutadiene, butylether, neoprene, polyisobutylene, polybutadiene, and
polyisoprene;
polyvinylacetate; unreaformaldehyde resins; phenolformaldehyde resins;
resorcinol
formaldehyde resins, cellulose derivatives such as ethylcellulose,
methylcellulose, nitrocellulose,
cellulose acetatebutyrate, and carboxymethyl cellulose; and natural gums such
as guar, acacia,
pectins, starch, dextrin, albumin, gelatin, casein, etc. The adhesives can be
compounded with
tackifiers and stabilizers, as is well known in the art.

Representative silicone adhesives include silicone elastomers based on
monomers of silanes,
halosilanes, or CMS alkoxysilanes, especially polydimethylsiloxanes which may
be used alone or
formulated with a silicone tackifier or silicone plasticizer which are
selected from medically
acceptable silicone fluids, i.e. non-elastomeric silicones based on silanes,
halosilanes or C1_i8
alkoxysilanes. Typical silicone adhesives are available from Dow Coming under
the tradename
SILASTIC .

Liquid Vehicles
Transdermal compositions can include a variety of components, including a
liquid vehicle,
typically a C2 alkanol such as ethanol, isopropanol, n-propanol, butanol, a
polyalcohol or glycol
such as propylene glycol, butylene glycol, hexylene glycol, ethylene glycol,
and/or purified
water. The vehicle is typically present in an amount of between about 5 and
about 75% w/w,


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
more typically, between about 15.0% and about 65.0% w/w, and, preferably,
between about 20.0
and 55.0% w/w.

Water augments the solubility of hydrophilic active agents in the composition,
and accelerates the
5 release of lipophilic active agents from a composition. Alcohols, such as
ethanol, increase the
stratum corneum liquid fluidity or function to extract lipids from the stratum
comeum. As
discussed herein, the glycols can also act as permeation enhancers.

Controlled Release of the Active Agent
10 The administration of the active agent can be controlled by using
controlled release compositions,
which can provide rapid or sustained release, or both, depending on the
compositions. There are
numerous particulate drug delivery vehicles known to those of skill in the art
which can include
the active ingredients, and deliver them in a controlled manner. Examples
include particulate
polymeric drug delivery vehicles, for example, biodegradable polymers, and
particles formed of
15 non-polymeric components. These particulate drug delivery vehicles can be
in the form of
powders, microparticles, nanoparticles, microcapsules, liposomes, and the
like. Typically, if the
active agent is in particulate form without added components, its release rate
depends on the
release of the active agent itself. In contrast, if the active agent is in
particulate form as a blend of
the active agent and a polymer, the release of the active agent is controlled,
at least in part, by the
20 removal of the polymer, typically by dissolution or biodegradation.

In one embodiment, the transdermal compositions can provide an initial rapid
release of the
active ingredient followed by a sustained release of the active ingredient.
U.S. Patent No.
5,629,011 provides examples of this type of composition. There are numerous
transdermal
25 compositions that use transdermal delivery to deliver nicotine in a time-
release manner (such as
rate-controlling membranes), including currently marketed nicotine replacement
therapies. These
are also suitable for administering the compounds described herein.

Semi-Solid Dosage Forms
30 In one embodiment, the transdermal dosage form is not a "patch," but
rather, a semisolid dosage
form such as a gel, cream, ointment, liquid, etc. In this embodiment, one can
augment patient's
compliance and cover a broader surface area than can be covered with a patch.


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
36
In this embodiment, particularly when used for pain treatment, the dosage form
can include other
active and inactive components typically seen in semisolid dosage forms used
to treat pain. These
include, but are not limited to, menthol, wintergreen, capsaicin, aspirin,
NSAIDs, narcotic agents
(e.g. fentanyl), alcohols, oils such as emulsion oil, and solvents such as
DMSO.
Iontophoresis
In addition to delivery via transdermal drug delivery devices and semi-solid
dosage forms, the
active ingredients can also be delivered via iontophoresis. Iontophoresis is a
non-invasive method
of propelling high concentrations of a charged substance, such as the active
ingredients described
herein, transdermal^ by repulsive electromotive force. The technique involves
using a small
electrical charge applied to an iontophoretic chamber containing a similarly
charged active agent
and its vehicle. The skin's permeability is altered upon application of the
charge, and this
increases migration of the active ingredient into the epidermis.

Iontophoresis can be used to transdermally deliver the active agents, using
active transportation
within an electric field, typically by electromigration and electroosmosis.
These movements are
typically measured in units of chemical flux, commonly mol/cm *h. The
isoelectric point of the
skin is approximately 4. Under physiological conditions, where the surface of
the skin is buffered
at or near 7.4, the membrane has a net negative charge, and electroosmotic
flow is from anode (-)
to cathode (+). Electroosmosis augments the anodic delivery of the (positively
charged) active
agents described herein.

Iontophoresis devices include two electrodes, which are typically attached to
a patient, each
connected via a wire to a microprocessor controlled electrical instrument. The
active agents are
placed under one or both of the electrodes, and are delivered into the body as
the instrument is
activated.

Typically, ions are delivered into the body from an aqueous drug reservoir
contained in the
iontophoretic device, and counter ions of opposite charge are delivered from a
"counter
reservoir." Solutions containing the active ingredient, and also solutions of
the counter ions, can
be stored remotely and introduced to an absorbent layer of the iontophoresis
electrode at the time
of use. Examples of such systems are described in U.S. Pat. Nos. 5,087,241;
5,087,242;
5,846,217; and 6,421,561, the contents of which are hereby incorporated by
reference.
Alternatively, as described in U.S. Patent No. 5,685,837, the active agents
can be pre-packaged in


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
37
dry form into the electrode(s). This approach requires a moisture activation
step at the time of
use.

Solutions of the active agents can be co-packaged with the iontophoretic
device, ideally
positioned apart from the electrodes and other metallic components until the
time of use. This
technique, and suitable devices, are described, for example, in U.S. Patent
Nos. 5,158,537;
5,288,289; 5,310,404; 5,320,598; 5,385,543; 5,645,527; 5,730,716; and
6,223,075. In these
devices, a co-packaged electrolyte constituent liquid is stored remotely from
the electrodes, in a
rupturable container and a mechanical action step at the time of use induces a
fluid transfer to a
receiving reservoir adjacent to the electrodes. These systems enable precise
fluid volumes to be
incorporated at the time of manufacture to avoid overfilling.

In addition to solutions, the active agents can be present in a pre-formed
gel, as described in U.S.
Patent No. 4,383,529, incorporated by reference. Thus, a preformed gel
containing the active
agent can be transferred into an electrode receptacle at the time of use. This
system can be
advantageous in that it provides a precise pre-determined volume of the gel,
thus preventing over-
filling. Further, since the active agent is present in a gel composition, it
is less likely to leak
during storage or transfer.

In some embodiments, the transdermal drug delivery is carried out using
devices that include a
polymeric barrier, adhered to the skin with a suitable adhesive, and which
also include a suitable
amount of the active ingredients, or salts thereof, in solution or dispersion
and in contact with the
skin or a rate-controlling membrane may be used between the active-containing
composition and
the skin. In others, the delivery is carried out using semisolid compositions,
such as cremes or
lotions, which include the active ingredients, and which are applied to the
skin. In still other
embodiments, the active ingredients are delivered using iontophoresis, wherein
the positively
charged active agents are administered by electroosmosis. There may also be
embodiments
wherein the active ingredient(s) is formulated within the matrix of the
adhesive.

As previously indicated, the present invention provide transdermal
compositions of (4aR,l0aR)-
1-n-propyl-1,2,3,4,4a,5,10,10a-octahydro-benzo[g]quinoline-6,7-diol and
related compounds,
which may be delivered to the systemic circulation via delivery across the
skin.


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
38
In one embodiment, the composition is further characterized as patch. In one
embodiment, the
composition is further characterized as a semisolid dosage form. In one
embodiment, the
composition is further characterized as a gel, lotion or creme. In one
embodiment, the
composition is further characterized as a controlled release formulation. In
one embodiment, the
composition is further comprising a permeation enhancer. In one embodiment,
the composition
is further comprising one or more adhesive, binder, lubricant, glidant,
disintegrant or mixture
thereof

The compound of the invention as a pharmaceutical composition for transdermal
adminstration
may be administered in any suitable way across the skin, and the compound may
be presented in
any suitable dosage form for such administration, e.g. in form of simple
solutions or dispersions,
simple tablets, matrix tablets, capsules, powders, syrups, dissolvable films,
patches, lipophilic
gels. In another embodiment, the compound of the invention is administered in
the form of a
dissolvable film.
In a specific embodiment of the invention, there is provided a transdermal
composition
comprising a therapeutically effective amount of the compound of the
invention, or a
pharmaceutically acceptable acid addition salt thereof, for administration
across the skin.

In a further aspect the invention provides the use of said composition for the
preparation of a
medicament for the treatment of neurodegenerative disorders such as
Parkinson's disease and
Huntington's disease.

In a further aspect the invention provides the use of the transdermal
composition for the
preparation of a medicament for the treatment of psychoses, impotence, renal
failure, heart failure
or hypertension.

In another aspect the invention provides the use of the transdermal
composition for the
manufacture of a medicament for the treatment of cognitive impairment in a
mammal.
In a still further aspect the invention provides the use of the transdermal
composition for the
manufacture of a medicament for the treatment of restless legs syndrome (RLS)
or periodic limb
movement disorder (PLMD).


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
39
In a still further aspect the invention provides the use of the transdermal
composition for the
manufacture of a medicament for the treatment of erectile dysfunction.

In a different aspect the invention provides the use of the transdermal
composition for the
manufacture of a medicament for the treatment of movement disorders, poverty
of movement,
dyskinetic disorders, gait disorders or intention tremor in a mammal.

In a further aspect the invention provides the use of the transdermal
composition for the treatment
of neurodegenerative disorders such as Parkinson's disease and Huntington's
disease.
In a further aspect the invention provides the use of the transdermal
composition for the treatment
of psychoses, impotence, renal failure, heart failure or hypertension.

In another aspect the invention provides the use of the transdermal
composition for the treatment
of cognitive impairment in a mammal.

In a still further aspect the invention provides the use of the transdermal
composition for the
treatment of restless legs syndrome (RLS) or periodic limb movement disorder
(PLMD).

In a different aspect the invention provides the use of the transdermal
composition for the
treatment of movement disorders, poverty of movement, dyskinetic disorders,
gait disorders or
intention tremor in a mammal.

In separate aspects the invention provides the use of the transdermal
composition for the
manufacture of medicaments, which are intended for administration via the
skin.

The invention also provides a method of treating a mammal suffering from a
neurodegenerative
disorder such as Parkinson's disease and Huntington's disease comprising
administering to the
mammal a therapeutically effective amount of the transdermal composition.
In another aspect the invention also provides a method of treating a mammal
suffering from
psychoses, impotence, renal failure, heart failure or hypertension, comprising
administering to the
mammal a therapeutically effective amount of the transdermal composition.


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
In a further aspect the invention provides a method of treating a mammal
suffering from a
cognitive impairment, comprising administering to the mammal an effective
amount of the
transdermal composition.

5 The invention also relates to a method of treating a mammal suffering from
restless legs
syndrome (RLS) or periodic limb movement disorder (PLMD), comprising
administering to the
mammal a transdermal composition of the compound of the invention, or a
pharmaceutically
acceptable addition salt thereof

10 The invention also relates in a separate aspect to a method of treating a
mammal suffering from
movement disorders, poverty of movement, dyskinetic disorders, gait disorders
or intention
tremor comprising administering to the mammal of the pharmaceutical
composition.

The therapeutically effective amount of the compound of the invention,
calculated as the daily
15 dose of the compound of the invention above as the free base, is suitably
between 0.001 and 12.5
mg/day, more suitable between 0.005 and 10.0 mg/day, e.g. preferably between
0.01 and 5.0
mg/day. In a specific embodiment the daily dose of the compound of the
invention is between 0.1
and 1.0 mg/day.

20 In another embodiment the daily dose of the compound of the invention is
less than about 0.1
mg/day. In a separate embodiment the daily dose of the compound of the
invention is about 0.01
mg/day. In a further embodiment the invention provides a formulation
comprising from 0.0001
mg to 12.5 mg of the compound of the invention for transdermal delivery. In a
further
embodiment the invention provides a formulation comprising from 0.0001 mg to
0.01 mg of the
25 compound of the invention for transdermal delivery. In a further embodiment
the invention
provides a formulation comprising from 0.001 mg to 0.10 mg of the compound of
the invention
for transdermal delivery. In a further embodiment the invention provides a
formulation
comprising from 0.01 mg to 1.0 mg of the compound of the invention for
transdermal delivery.

30 Ultimately, the exact dose of the compound of the invention and the
particular formulation to be
administered depend on a number of factors, e.g., the condition to be treated,
the desired duration
of the treatment and the rate of release of the active agent. For example, the
amount of the active
agent required and the release rate thereof may be determined on the basis of
known in vitro or in


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
41
vivo techniques, determining how long a particular active agent concentration
in the blood plasma
remains at an acceptable level for a therapeutic effect.

Pharmaceutically Acceptable Salts of Compound 10
Compound 10 and related compounds form pharmaceutically acceptable acid
addition salts
with a wide variety of organic and inorganic acids. Such salts are also part
of this invention.
A pharmaceutically acceptable acid addition salt of the compound of the
invention is formed
from a pharmaceutically acceptable acid as is well known in the art. Such
salts include the
pharmaceutically acceptable salts listed in Journal of Pharmaceutical Science,
66, 2-19
(1977) and are known to the skilled person. Typical inorganic acids used to
form such salts
include hydrochloric, hydrobromic, hydroiodic, nitric, sulphuric, phosphoric,
hypophosphoric, metaphosphoric, pyrophosphoric, and the like. Salts derived
from organic
acids, such as aliphatic mono and dicarboxylic acids, phenyl substituted
alkanoic acids,
hydroxyalkanoic and hydroxyalkandioic acids, aromatic acids, aliphatic and
aromatic
sulfonic acids, may also be used. Such pharmaceutically acceptable salts thus
include the
chloride, bromide, iodide, nitrate, acetate, phenylacetate, trifluoroacetate,
acrylate, ascorbate,
benzoate, chlorobenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate,
methylbenzoate, o-acetoxybenzoate, isobutyrate, phenylbutyrate,
hydroxybutyrate, butyne-
1,4-dicarboxylate, hexyne-l,4-dicarboxylate, caprate, caprylate, cinnamate,
citrate, formate,
fumarate, glycollate, heptanoate, hippurate, lactate, malate, maleate,
hydroxymaleate,
malonate, mandelate, mesylate, nicotinate, isonicotinate, oxalate, phthalate,
teraphthalate,
propiolate, propionate, phenylpropionate, salicylate, sebacate, succinate,
suberate,
benzenesulfonate, p-bromobenzenesulfonate, chlorobenzenesulfonate,
ethylsulfonate, 2-
hydroxyethylsulfonate, methylsulfonate, naphthalene- l-sulfonate, naphthalene-
2-sulfonate,
naphthalene- 1,5-sulfonate, p-toluenesulfonate, xylenesulfonate, tartrate, and
the like.


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
42
BRIEF DESCRIPTION OF THE FIGURES

FIGURE 1: Crystal structure of compound ent-10. The absolute configuration was
determined
by the anomalous scattering of the `heavy' bromine atom.

FIGURE 2: Dose-response curve for the concentration-dependent stimulation of
intracellular
Cat release by dopamine in hD5-transfected CHO-Gal6 cells.

FIGURE 3: Representative Chromatogram of Sample from animal 2, Day 5
FIGURE 4: Dose Normalised AUCO-00 for Compound 10 from Example 14
FIGURE 5: Dose Normalised Cmax for Compound 10 from Example 14


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
43
EXPERIMENTAL SECTION

Analytical LC/MS data were obtained on a PE Sciex API 150EX instrument
equipped with
atmospheric pressure photo ionization and a Shimadzu LC-8A/SLC-1OA LC system.
Purity was
determined by integration of the UV (254 nm) and ELSD traces. MS instruments
are from
Peskier (API), equipped with APPI-source and operated in positive ion mode.
The retention times
in the UV-trace (RT) are expressed in min. Solvents A was made of 0.05% TFA in
water, while
solvent B was made of 0.035% TFA and 5% water in acetonitrile. Several
different methods have
been used:

Method 25: API 150EX and Shimadzu LC10AD/SLC-1OA LC system. Column: dC-18
4.6x30mm, 3 microm (Atlantis, Waters). Column temperature: 40 C. Gradient:
reverse phase
with ion pairing. Flow: 3.3 mL/min. Injection volume: 15 microL. Gradient: 2%
B in A to 100%
B over 2.4 min then 2% B in A for 0.4 min. Total run time: 2.8 min.

Method 14: API 150EX and Shimadzu LC8/SLC-1OA LC system. Column: C-18
4.6x30mm,
3.5microm (Symmetry, Waters). Column temperature: rt. Gradient: reverse phase
with ion
pairing. Flow: 2mL/min. Injection volume: 10 microL. Gradient: 10% B in A to
100% B over 4
min then 10% B in A for 1 min. Total run time: 5 min.

X-ray crystal structure determination was performed as follows. The crystal of
the compound
was cooled to 120 K using a Cryostream nitrogen gas cooler system. The data
were collected on
a Siemens SMART Platform diffractometer with a CCD area sensitive detector.
The structures
were solved by direct methods and refined by full-matrix least-squares against
F2 of all data. The
hydrogen atoms in the structures could be found in the electron density
difference maps. The
non-hydrogen atoms were refined anisotropically. All the hydrogen atoms were
at calculated
positions using a riding model with O-H=0.84, C-H = 0.99-1.00, N-H = 0.92-0.93
A. For all
hydrogen atoms the thermal parameters were fixed [U(H) = 1.2 U for attached
atom]. The Flack
x-parameters are in the range 0.0(1)-0.05(1), indicating that the absolute
structures are correct.
Programs used for data collection, data reduction and absorption were SMART,
SAINT and
SADABS [cf. "SMART and SAINT, Area Detector Control and Integration Software",
Version
5.054,Bruker Analytical X-Ray Instruments Inc., Madison, USA (1998), Sheldrick
"SADABS,
Program for Empirical Correction of Area Detector Data" Version 2.03,
University of Gottingen,
Germany (2001)]. The program SHELXTL [cf. Sheldrick "SHELXTL, Structure
Determination


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
44
Programs", Version 6.12, Bruker Analytical X-Ray Instruments Inc., Madison,
USA (2001)] was
used to solve the structures and for molecular graphics.

Synthesis of the compounds of the invention

Starting from compound 1 whose synthesis is described in the literature
prepared as described in
Taber et at., J. Am. Chem. Soc., 124(42), 12416 (2002), compound 8 can be
prepared as
described herein in eight steps. This material can be resolved by chiral SFC
as described herein to
give compounds 9 and ent-9. After cleavage of the Boc-protective group,
reductive amination can
be used to introduce the n-propyl group on the nitrogen atom. The resulting
masked catechol
amines can be deprotected under standard conditions by treatment with 48% HBr
or by reaction
with BBr3 to give compounds 10 and ent-10.

The enantiomer of example 1 (compound 10) and ent-example 1 (ent-compound 10),
can be
prepared in a similar manner from ent-9. The racemate of example 1, rac-
example 1, can be
prepared by mixing a 1:1 mixture of example 1 and ent-example 1. It can also
be obtained from
non-resolved compound 8 or a 1:1 mixture of compound 9 / ent-9 as described
above for the pure
enantiomers. Alternatively, rac-example 1 can be prepared as described in the
literature (Cannon
et al., J. Heterocycl. Chem. 17, 1633 (1980)).

0 10
.OMe
eight steps

MeO MeO' D
We We
compound 1 compound 8
(racemate)
SFC
(resolution)

O~O~ O-Y O-~
MeO~~ Meoj~ 1
We We
compound 9 ent-compound 9
(4aR, lOaR-enantiomer) (4aS, lOaS-enantiomer)

HO J / HO HO D
OH OH OH
compound 10 ent-compound 10 rac-compound 10
(4aR, lOaR-enantiomer) (4aS,10aS-enantiomer)


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
Synthesis of compounds 9 and ent-9.

7-Iodo-1,2,6-trimethoxy-naphthalene (compound 2).

:aOMe OMe
Me0 I MeO I I
OMe OMe
compound 1 compound 2
5 To a stirred solution of compound 1 (26.2 g; prepared as described in Taber
et at., J. Am. Chem.
Soc., 124(42), 12416 (2002)) in dry THE (200 mL) under argon and at -78 C was
slowly added
s-butyl lithium (1.2 M in cyclohexane, 110 mL). The solution was stirred at -
78 C for 3h. A
solution of iodine (30.5 g) in dry THE (50 mL) was added over a period of 10
min. The resulting
mixture was then stirred for another 10 min at -78 C. The reaction mixture
was quenched by the
10 addition of sat. NH4C1 (100 mL), water (240 mL), and Et20 (240 mL). The
organic layer was
washed with 10% aqueous sodium sulfite solution (100 mL), dried (Na2SO4) and
concentrated in
vacuo. The crude material was purified by distilling off unreacted starting
material. The residue
was further purified by silica gel chromatography (EtOAc/heptane) to produce
an impure solid
material, which was purified by precipitation from EtOAc/heptane affording
11.46 g of
15 compound 2.

(E/Z)-3-(3,7,8-Trimethoxy-naphthalen-2-yl)-acrylonitrile (compound 3).

OMe - I OMe
MeO MeO CN
OMe OMe
compound 2 compound 3
To a suspension of compound 2 (3.41 g) in dry acetonitrile (10.7 mL) in a
microwave reactor vial
20 was added acrylonitrile (1.19 mL) Pd(OAc)2 (73 mg), and triethylamine (1.48
mL). The vial was
sealed, and the mixture was heated for 40 min at 145 C under microwave
irradiation. This
procedure was carried out two more times (using a total of 10.23g of compound
5). The crude
reaction mixtures were combined and the catalyst was filtered off, and the
filtrate was
concentrated in vacuo. The residue was partitioned between Et20 (300 ML) and
2M HC1 (150
25 mL). The organic layer was washed with brine (100 mL), dried (Na2SO4) and
concentrated in
vacuo. The crude material (7.34 g) was purified by silica gel chromatography
(EtOAc/heptane) to
produce 5.23 g of compound 3 as a mixture of olefin isomers.


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
46
3-(3,7,8-Trimethoxy-naphthalen-2-yl)-propionitrile (compound 4).
OMe OMe
MeO I CN MeO CN
OMe OMe
compound 3 compound 4
Compound 3 (5.23 g) was dissolved in CHC13 (15 mL) and 99% EtOH (100 mL). 10%
Pd/C (0.8
g) was added and the solution was hydrogenated for 45 min under a hydrogen
pressure of 3 bar
using a Parr shaker. The catalyst was filtered off, and the filtrate was
passed through a small
plough of silica gel (eluent: 99% EtOH). Yield: 4.91 g compound 4 as a white
solid.

[3-(3,7,8-Trimethoxy-1,4-dihydro-naphthalen-2-yl)-propyl]-carbamic acid t-
butyl ester
(compound 5).

T
~ OMe _ OMe OO
Me0 I CN Me0 I I NH
OMe OMe
compound 4 compound 5
Compound 4 (5.0g) was dissolved in 99% EtOH (150 mL) and the mixture was
heated to reflux
under nitrogen atmosphere. Sodium metal (5g) was added in small lumps over 3h.
The mixture
was refluxed for an additional 2h, before it was stirred at rt for 2 days.
Then it was heated to
reflux again, and more sodium metal (3.68 g) was added and the mixture was
refluxed overnight.
After cooling on an ice/water bath, the reaction was quenched by the addition
of solid ammonium
chloride (20 g) and water (25 mL). The resulting mixture was filtered, and the
filtrate was
concentrated in vacuo. The residue was partitioned between diethyl ether (50
mL) and water (50
mL). The aqueous layer was neutralized with 37% HC1 and extracted with diethyl
ether (2x50
mL). The combined organic extracts were washed with brine (50 mL), dried
(MgS04) and
concentrated in vacuo to afford an oil. This material was dissolved in THE (50
mL) and treated
with Boc2O (2.34 g) and Et3N (1.78 mL) at rt. After six days the volatiles
were removed in vacuo
and the residue was purified by silica gel chromatography (EtOAc/heptane).
This provided
impure compound 5 (1.52 g).


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
47
Racemic 6,7-dimethoxy-2,3,4,4a,5,10-hexahydro-benzo[g]quinoline hydrochloride
(compound 6).

OMe 0 %HCI
MeO I I NH Me0 JIC.4; OMe OMe
ompound 5 compound 6
(racemate)

Compound 5 (1.52 g from the previous step) was dissolved in MeOH (20 mL). 37%
HC1 (3.5
mL) was added, and the mixture was refluxed for 4h. The volatiles were removed
in vacuo, using
toluene to azeotropically remove the water. This provided impure compound 6
(0.89 g) as an
yellow oil.

Racemic trans-6,7-dimethoxy-3,4,4a,5,10,10a-hexahydro-2H-benzo[g]quinoline-l-
carboxylic acid t-butyl ester (compound 8).

HCI H HCI 0 yo-f~
Me0 I MeO I MeO
OMe OMe OMe
compound 6 compound 7 compound 8
(racemate) (racemate) (racemate)
Compound 6 (0.89 g) was dissolved in MeOH (10 mL) and NaCNBH3 (0.19 g) was
added. The
reaction was stirred overnight at rt. The crude mixture was cooled on an
ice/water bath, before it
was quenched with 2 M HC1 in Et20 (1 mL). The mixture was partitioned between
Et20 (50
mL), water (50 mL), and 2 M NaOH (10 mL). The aqueous layer was extracted with
diethyl ether
(3x50 mL). The combined organic layers were dried (MgSO4) and concentrated in
vacuo to
afford the impure free amine (compound 7). This material was dissolved in THE
(25 mL) and
treated with Boc2O (0.68 g) and Et3N (0.86 mL) at rt for lh. The crude mixture
was concentrated
in vacuo, and the residue was purified by silica gel chromatography
(EtOAc/heptane) to provide
1. l 8g of racemic compound 8 sufficiently pure for the next step.

SFC-separation of the enantiomers of racemic trans-6,7-dimethoxy-
3,4,4a,5,10,10a-
hexahydro-2H-benzo[g]quinoline-l-carboxylic acid t-butyl ester (compounds 9
and ent-9).

t
MeO MeO 9 MeO
OMe OMe OMe
compound 8 compound 9 compound ent-9
(racemate) (4R,1OR enantiomer) (4S,10S enantiomer)


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
48
Compound 8 (19.7 g) was resolved into its enantiomers using chiral SFC on a
Berger SFC
multigram II instrument equipped with a Chiralcel OD 21.2 x 250 mm column.
Solvent system:
C02/EtOH (85:15), Method: constant gradient with a flow rate of 50 mL/min.
Fraction collection
was performed by UV 230 nm detection. Fast eluting enantiomer (4aR, lOaR
enantiomer;
compound 9): 9.0 g of a white solid. Slow eluting enantiomer (4aS, l OaS
enantiomer; compound
ent-9): 8.1 g of a white solid.

(4aS,10aS)-6,7-Dimethoxy-1,2,3,4,4a,5,10,10a-octahydro-benzo[g]quinoline
hydrochloride
(compound ent-9').
O O~
N H HCI
N
MeO I / I /
Me0
OMe OMe
compound ent-9 compound ent-9'
(4S, 10S enantiomer) (4S, 1OS enantiomer)
Compound ent-9 (0.52g) was dissolved in MeOH (15 mL) and treated with 5 M HC1
in Et20 (7.5
mL) at rt for 2h. . The mixture was concentrated in vacuo and the solid was
dried in vacuo to give
compound ent-9' as a white solid. LC/MS (method 14): RT 1.31 min.

Example 1. Preparation of the compounds of the invention
Synthesis of (4aR,1OaR)-1-n-Propyl-1,2,3,4,4a,5,10,10a-octahydro-
benzo[g]quinoline-6,7-
diol hydrobromide (compound 10).

O_` 'O ~Br
MeO (/ ~`)
lOMe OH
compound 9 compound 10
(4aR, 1OaR enantiomer) (4aR, 1OaR enantiomer)
Compound 9 (0.5 g) was dissolved in 99% EtOH (5 mL) and treated with 2M HC1 in
Et20 (4
mL) overnight at rt. The crude mixture was concentrated in vacuo, and the
residue was
partitioned between EtOAc and 10% aqueous NaOH (5 mL). The aqueous layer was
extracted
with EtOAc, and the combined organic layers were washed with brine, dried
(MgS04),
concentrated in vacuo. The residue was dissolved in 99% EtOH (5 mL) and
treated with
propionic aldehyde (0.52 mL), NaCNBH3 (0.45 g), and AcOH (3 drops) overnight
at rt. The
crude mixture was portioned between sat. aqueous NaHCO3 (12.5 mL), water (12.5
mL), and
EtOAc (2x25 mL). The combined organic layers were washed with brine, dried
(MgS04), and
concentrated in vacuo. The residue was purified by silica gel chromatography
(MeOH/EtOAc).


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
49
The obtained intermediate was treated with 48% HBr (3 mL) at 150 C for lh
under microwave
conditions, before the crude mixture was stored at 4 C overnight. The
precipitated material was
isolated by filtration and dried in vacuo. Yield of compound 10: 103 mg as a
solid. LC/MS
(method 25): RT 0.77 min.

(4aS,lOaS)-1-Propyl-1,2,3,4,4a,5,10,10a-octahydro-benzo [g] quinoline-6,7-diol
hydrobromide (compound ent-10).

.H HCI pN ~{Br
Me0 I O
OMe OH
compound ent-9' compound ent-10
(4aS, 1 OaS enantiomer) (4aS, 10aS enantiomer)
The procedure described for compound 10 was followed starting from compound
ent-9' (0.5 g;
the HC1 salt was liberated by partitioning between EtOAc and 10% aqueous NaOH
before the
reductive amination step). Yield of compound ent-10: 70 mg as a solid. LC/MS
(method 25): RT
0.70 min. A small sample of compound ent-10 was dissolved in MeOH and allowed
to crystallize
slowly at rt over 2 months. The formed white crystals were collected and
subjected to X-ray
analysis (cf. Figure 1). The absolute configuration of compound ent-10 was
determined by X-ray
crystallography and allowed for unambiguous determination of the
stereochemistry of
compounds 9 and 10 and hence their related compounds.

Example 2. General Diester syntheses
The scheme below provides a general procedure for the conversion of
catecholamines to the
symmetric, asymmetric and mono esters of compound 10.

HO / C
HO ' = ."=,~ R~F:c0 O O O O / )

OH Ry OH yR~
O O
Formula la Formula lb Formula lc

wherein each R,,, Ry, and Rz is independently C1.6 alkanoyl, cycloalkylalkyl,
phenylacetyl or
benzoyl, or a pharmaceutically acceptable salt thereof, and a pharmaceutically
acceptable carrier.
Briefly, the catechol amine was treated with acylchloride using TFA as
solvent. The crude acyl
catecholamine(s) was purified by aluminum oxide chromatography (for a
reference on this
transformation, see for example: Wikstrom, Dijkstra, Cremers, Andren,
Marchais, Jurva; WO


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
02/14279). Each of the symmetric, asymmetric and mono-esters described in this
example falls
within the scope of this invention.

Example 3: 2,2-Dimethyl-propionic acid (4aR,10aR)-7-(2,2-dimethyl-
5 propionyloxy)-1-n-propyl-1,2,3,4,4a,5, 10,10a-octahydro-benzo[g]quinolin-6-
yl ester
trifluoroacetate
As a working example, but without limiting the scope of the subject invention,
a symmetrical
diester was prepared in a similar manner as described above starting from
compound 10 (44 mg)
and pivaloyl chloride. Yield of Example 3 was 14 mg as a white solid. LC/MS
(method 14): RT
10 2.45 min, ELSD 97.7%, UV 83.9%. MH 430.2.

Pharmacological data

Example 4: Pharmacological Testing in vitro I
D1 cAMP assay
15 The ability of the compounds to either stimulate or inhibit the D1 receptor
mediated cAMP
formation in CHO cells stably expressing the human recombinant D1 receptor was
measured as
follows. Cells were seeded in 96-well plates at a concentration of 11000
cells/well 3 days prior to
the experiment. On the day of the experiment the cells were washed once in
preheated G buffer
(1 mM MgC12, 0.9 mM CaC12, 1 mM IBMX (3-i-butyl-l-methylxanthine) in PBS
(phosphate
20 buffered saline)) and the assay was initiated by addition of 100 micro-L of
a mixture of 30 nM
A68930 and test compound diluted in G buffer (antagonism) or test compound
diluted in G
buffer (agonism).

The cells were incubated for 20 minutes at 37 C and the reaction was stopped
by the addition of
100 micro-L S buffer (0.1 M HC1 and 0.1 mM CaC12) and the plates were placed
at 4 C for lh.
25 68 micro-L N buffer (0.15 M NaOH and 60 mM NaOAc) was added and the plates
were shaken
for 10 minutes. 60 micro-1 of the reaction were transferred to cAMP
FlashPlates (DuPont NEN)
containing 40 micro-L 60 mM Sodium acetate pH 6.2 and 100 micro-L IC mix (50
mM Sodium
acetate pH 6.2, 0.1 % sodium azide, 12 mM CaC12, 1% BSA (bovine serum albumin)
and 0.15
micro-Ci/mL 125I-CAMP) were added. Following an 18h incubation at 4 C the
plates were

30 washed once and counted in a Wallac TriLux counter. Compound 10 was
demonstrated to act as
a D1 agonist in this assay with an EC50 of 15.5 nM and an intrinsic activity
(efficacy) of 100%. In


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
51
comparison, apomorphine and dopamine were D1 agonists in this assay with EC50-
values of 52
nM and 43 nM, respectively and intrinsic activities (efficacies) of 86% and
100%, respectively.
Example 5: Pharmacological Testing in vitro II

D2 cAMP assay
The ability of the compounds to either stimulate or inhibit the D2 receptor
mediated inhibition of
cAMP formation in CHO cells transfected with the human D2 receptor was
measured as follows.
Cells were seeded in 96 well plates at a concentration of 8000 cells/well 3
days prior to the
experiment. On the day of the experiment the cells were washed once in
preheated G buffer (1

mM MgC12, 0.9 mM CaC12, 1 mM IBMX in PBS) and the assay was initiated by
addition of 100
micro-1 of a mixture of 1 micro-M quinpirole, 10 microM forskolin and test
compound in G
buffer (antagonism) or 10 micro-M forskolin and test compound in G buffer
(agonism).

The cells were incubated 20 minutes at 37 C and the reaction was stopped by
the addition of 100
microL S buffer (0.1 M HCl and 0.1 mM CaC12) and the plates were placed at 4
C for lh. 68
microL N buffer (0.15 M NaOH and 60 mM Sodium acetate) were added and the
plates were
shaken for 10 minutes. 60 micro-L of the reaction were transferred to cAMP
FlashPlates (DuPont
NEN) containing 40 micro-L 60 mM NaOAc pH 6.2 and 100 micro-L IC mix (50 mM
NaOAc
pH 6.2, 0.1 % Sodium azide, 12 mM CaC12, 1% BSA and 0.15 micro-Ci/ml 125I-
cAMP) were

added. Following an 18h incubation at 4 C the plates were washed once and
counted in a Wallac
TriLux counter. Compound 10 was demonstrated to act as a D5 agonist in this
assay with an EC50
of 0.11 nM and an intrinsic activity (efficacy) of 100%. In comparison,
apomorphine and
dopamine were D2 agonists in this assay with EC50-values of 3.9 nM and 21 nM,
respectively and
intrinsic activities (efficacies) of 100% for both compounds.
Example 6: Pharmacological Testing in vitro III
D5 assay
Concentration-dependent stimulation of intracellular Ca2+ release by dopamine
in hD5-transfected
CHO-Ga16 cells. The cells were loaded with fluoro-4, a calcium indicator dye,
for 1h. Calcium
response (fluorescence change) was monitored by FLIPR (fluorometric imaging
plate reader) for
2.5 min. Peak responses (EC50) were averaged from duplicate wells for each
data point and
plotted with drug concentrations (cf Figure 2 for dopamine). Compound 10 was
demonstrated to
act as a D5 agonist in this assay with an EC50 of 0.06 nM and an intrinsic
activity (efficacy) of


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
52
95%. In comparison, apomorphine and dopamine were D5 agonists in this assay
with EC50-values
of 0.36 nM and 1.6 nM, respectively and intrinsic activities (efficacies) of
88% and 100%,
respectively.

Example 7: Pharmacological Testing in vivo I
D1/D2 dissections
Dopamine agonists can have activity at either the Dl receptors, the D2
receptors, or both. We
have used the rotation response in rats with unilateral 6-OHDA lesions to
assess compounds for
their ability to stimulate both receptor types and induce rotation [Ungerstedt
and Arbuthnott,
Brain Res. 1970, 24, 485; Setter et at., Eur. J. Pharmacol. 1978, 50(4), 419;
and Ungerstedt et at.
"Advances in Dopamine Research" (Kohsaka, Ed.), Pergamon Press, Oxford, p. 219
(1982)]. 6-
OHDA (6-hydroxydopamine) is a neurotoxin used by neurobiologists to
selectively kill
dopaminergic neurons at the site of injection in the brain in experimental
animals. In the 6-
OHDA model the nigrostraital dopamine cells are destroyed on one side of the
brain (unilateral)
by injecting 6-OHDA into the median forebrain bundle, located in front of the
substantia nigra.
The effects of the unilateral lesion combined with the administration of
dopamine agonists such
as apomorphine will induce rotation behaviour. Rats weighing 200-250 g were
subjected to
unilateral 6-OHDA lesions. Animals were permitted minimum three weeks to
recover before
being tested for rotation response to amphetamine (2.5 mg/kg subcutaneously)
and only animals
that responded by ipsolateral rotations were used in subsequent dyskinesia
studies (examples 8
and 9). Amphetamine increases dopamine levels in the synapse by blocking
reuptake and
increasing release from presynaptic terminals. This effect is greater in the
unlesioned side
causing the animals to rotate in the opposite direction as compared to their
response to direct
agonists such as L-DOPA and apomorphine that act predominantly on the lesioned
side of the
brain. For D1/D2 in vivo dissection studies were trained on apomorphine (0.1
mg/kg
subcutaneously) before being using in experiments and only animals that
repeatedly rotated at
least 350 times in 90 min were included. Rats where then randomly allocated to
the three
treatment groups balancing the groups for the animals' rotation response to
apomorphine (0.1
mg/kg subcutaneously). For dyskinesia studies animals were not trained on
apomorphine; instead
they were either primed with L-DOPA (example 9) or used `drug-naive' (example
8).
Experiments consist of determining a minimum effective dose (MED) to induce
rotation for the
compound in question. Once a MED has been determined, a second experiment is
performed to
determine the MED of the compound to overcome Nemonapride block
(MEDNemonapride).


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
53
Nemonapride is a D2 antagonist that blocks the D2 receptors, therefore any
observed rotations
would be dependent upon activity at the D 1 receptors. Finally, once the
MEDNemonapride is known
a third experiment is run using the MEDNemonapride dose and observing the
effect of the Dl
antagonist, SCH 23390 alone, the D2 antagonist, Nemonapride alone and finally,
the effect of
combined treatment with SCH 23390 and Nemonapride. This third experiment
confirms the
activity of the compound at both receptors as either antagonist alone can only
partially inhibit the
rotation response induced by the test compound while the combination treatment
completely
blocks all rotations in the rats [Arnt and Hyttel, Psychopharmacology, 1985,
85(3), 346; and
Sonsalla et at., J. Pharmacol Exp. Then., 1988, 247(1), 180]. This model was
validated using
Apomorphine as the proof-of-principle compound for mixed Dl/ D2 agonists.
Compound 10
(administered subcutaneously) had a mixed D1/D2 ratio of about 2 in this model
as compared to
apomorphine that had a ratio of about 3. A Dl component could not be observed
for D2-agonists
as exemplified by pramipexole and rotigotine. The data are summarized in Table
1.

Table 1. MED and MEDNemonapride for apomorphine, pramipexole, rotigotine, and
compound 10 (all compounds dosed SC).

apomorphine rotigotine pramipexole compound 10
MED 0.010 mg/kg 0.030 mg/kg 0.1 mg/kg 0.00065 mg/kg
MEDNemonapride 0.030 mg/kg 0.30 mg/kg* 1.0 mg/kg* 0.0013 mg/kg
*Rotations could not be blocked by administration of SCH23390.

Compound 10 has the in vivo profile of a long-lasting dual Dl/D2 agonist with
a fast onset of
action (when dosed buccally or s.c.). Thus, it would be expected that compound
10 could be
useful in treating ON/OFF fluctuations in Parkinson's Disease. It may also be
used as a 'rescue
drug' for the OFF periods (freezing).

Example 8: Pharmacological Testing in vivo II
Dyskinesia model with naive 6-OHDA rats
Twenty rats with unilateral 6-OHDA lesions [see example 7 for experimental
details] were used
to test induction of dyskinesia by compound 10 (administered subcutaneously;
n=7; group 1)
compared to L-DOPA/benserazide (6mg/kg / 15mg/kg subcutaneously; n=7; group 2)
and
apomorphine (lmg/kg subcutaneously; n=6; group 3). Benserazide is a DOPA
decarboxylase
inhibitor which is unable to cross the blood-brain barrier; it is used to
prevent metabolism of L-
DOPA to dopamine outside the brain.


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
54
During the actual dyskinesia experiments, rats received once daily injections
of the test
compounds subcutaneously and were observed for 3h following injection. Each
animal was
observed for 1 minute every 20 min throughout the 3h period for the presence
of dyskinesias
using the Abnormal Involuntary Movement Scale (AIMS) as described previously
(Lundblad et
at., Eur. JNeurosci., 15, 120(2002)). Rats received drug for 14 consecutive
days and were scored
on days 1, 2, 3, 4, 5, 8, 10 and 12. Two-way repeated measures ANOVA revealed
that there was
a significant treatment effect, time effect and treatment by time interaction
(p<O.001, in all cases).
Post hoc comparisons using Holm-Sidak method indicates that animals treated
with compound
10 had significantly less dyskinesia (scores of about 30) compared to animals
treated with either
L-DOPA or apomorphine (scores of about 65). There were no differences between
L-DOPA and
apomorphine treated groups. Following this experiment all rats were given
subcutaneous
injections of compound 10 from day 15-19 in order to determine how compound 10
influenced
the severity of dyskinesia seen in the apomorphine and L-DOPA groups.
Dyskinesia scoring was
performed on day 19 of the experiment (corresponding to 5 days on compound
10). The data
showed a partial reversal of the dyskinesias induced by L-DOPA and apomorphine
to about the
level of dyskinesias induced by compound 10 (which did not cause an increase
in dyskinesia in
group 1 as compared to the score of about 30 observed after 12 days of
treatment). The data are
presented in Table 2.
Table 2. Induction of dyskinesias by compound 10, L-DOPA, and apomorphine as
well as
reduction of dyskinesias induced by L-DOPA or apomorphine by treatment with
compound 10.
group 1 group 2 group 3
dose(once daily compound 10 L-DOPA / Benserazide apomorphine
on days 1-14) 0.0013 mg/kg SC 6/15 mg/kg SC 1 mg/kg SC
mean AIM score 27 66 61
(days 1 -12)
dose (once daily compound 10 compound 10 compound 10
on days 15 -19) 0.0013 mg/kg SC 0.0013 mg/kg SC 0.0013 mg/kg SC
mean AIM score 25 18 39
(day 19)

Example 9: Pharmacological Testing in vivo III
Reversal of L-DOPA-induced dyskinesias in 6-OHDA rats
A separate dyskinesia study addressed the reversal of L-DOPA induced
dyskinesias with either
pramipexole or Compound 10. Briefly, 18 animals were treated with L-
DOPA/Benserazide


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
(6/15mg/kg subcutaneously) for 7 days. Animals were observed on Days 1, 3 and
5 and AIMS
were scored. The day 5 scores were then used to separate the animals into
three groups of 6
animals each. Group 1 continued with daily L-DOPA treatment. Group 2 was
treated with
compound 10 (administered subcutaneously). Group 3 was treated with
pramipexole (0.16mg/kg
5 subcutaneously). Treatment continued daily for 10 days and the amount of
dyskinesia was
scored on days 1, 5, 9 and 10. Two-way repeated measures analysis of variance
indicated that
animals treated with compound 10 had significantly fewer dyskinesias than both
the pramipexole
group and the L-DOPA/Benserazide group. The pramipexole group had
significantly less
dyskinesias than the L-DOPA/Benserazide group. Hence, compound 10 had a
superior profile
10 over pramipexole in terms of reversing dyskinesias induced by L-DOPA. The
data are presented
in Table 3.

Table 3. Reduction of L-DOPA induced dyskinesias by treatment with compound 10
or
Pramipexole.

group 1 group 2 group 3
L-DOPA / compound 10
dose (once daily Benserazide 0.0013 mg/kg SC pramipexole
on days l -10) 6/15 mg/kg SC 0.16 mg/kg SC
mean AIM score 75 44 58
(days 1,5,9,10)
Accordingly, it is expected that dyskinesias in moderate to severe PD based on
L-DOPA-like
efficacy and reversal of dyskinesias can be treated by administration of
compound 10.

Example 10: Pharmacological Testing in vivo IV
Superiority model
Apomorphine and L-DOPA are able to reverse motility deficits in a mouse model
of severe
dopamine depletion. Both Apomorphine and L-DOPA stimulate Dl and D2 dopamine
receptors.
Pramipexole, an agonist at D2 receptors is ineffective in this model. Compound
10 has been
tested in this model and exhibits a profile similar to Apomorphine and L-DOPA
in that they are
able to restore locomotion in the mice. In this way, compound 10 is `superior'
to other
compounds, such as Pramipexole that target D2 receptors only. Bromocriptine is
another
example of a D2 agonist that does not reverse the deficits in this animal
model.


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
56
The experiments were performed as follows: Mice previously treated with MPTP
(2xl5mg/kg
subcutaneously) and that had stable lesions were used and vehicle treated mice
served as normal
controls. MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) is a neurotoxin
that causes
permanent symptoms of Parkinson's disease by killing certain neurons in the
substantia nigra of
the brain. It is used to study the disease in monkeys and mice. On the day of
the experiment, mice
were treated with AMPT (250mg/kg subcutaneously) and then returned to their
home cages for
1.5 hours after which they were placed in individual cages in the motility
unit. AMPT (alpha-
methyl-p-tyrosine) is a drug that temporarily reduces brain catecholamine
activity (in this case
especially dopamine levels). Three hours after the AMPT injection, rescue of
locomotive deficits
was attempted with compound 10 and activity was recorded for an additional 1.5
hours. The first
30 min of data collected after the rescue treatment was `contaminated' due to
stressing the
animals with handling and injection as evidenced by increased levels in the
vehicle controls
therefore the data were analyzed using the last 1 hour of recorded data.
Various compounds (all
dosed subcutaneously) were tested for their ability to reverse the motility
deficits produced in this
model. L-DOPA/Benserazide, apomorphine, and compound 10 restored locomotion in
the mice
in a dose-dependent manner. In contrast, the D2 agonists, pramipexole and
bromocriptine did not.
The data are presented in Tables 4a-4e.


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
57
Table 4a. L-DOPA/Benserazide reverses hypomotility in the MPTP/AMPT mouse
model.

AMPT AMPT
pretreatment (-1.5h) vehicle 250 mg/kg SC 250 mg/kg SC
L-DOPA and
treatment (0h) vehicle vehicle benserazide
50/50 mg/kg SC
activity count (0.5 -1.5h) 365 44 676
Table 4b. Apomorphine reverses hypomotility in the MPTP/AMPT mouse model.
AMPT AMPT
pretreatment (-1.5h) vehicle 250 mg/kg SC 250 mg/kg SC
treatment (0h) vehicle vehicle aomorphine
1.0 mg/kg SC
activity count (0.5 - 1.5h) 694 1 912

Table 4c. Compound 10 reverses hypomotility in the MPTP/AMPT mouse model
AMPT AMPT
pretreatment (-1.5h) vehicle 250 mg/kg SC 250 mg/kg SC
treatment (0h) vehicle vehicle compound 10
0.003 mg/kg SC
activity count (0.5 - 1.5h) 405 12 8
AMPT AMPT
pretreatment (-1.5h) 250 mg/kg SC 250 mg/kg SC
treatment (0h) compound 10 compound 10
0.01 mg/kg SC 0.03 mg/kg SC
activity count (0.5 -1.5h) 228 440


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
58
Table 4d. Bromocriptine does not reverse hypomotility in the MPTP/AMPT mouse
model.

AMPT AMPT
pretreatment (-1.5h) vehicle 250 mg/kg SC 250 mg/kg SC
treatment (0h) vehicle vehicle bromocriptine
l mg/kg SC
activity count (0.5 - 1.5h) 336 16 25
AMPT AMPT
pretreatment (-1.5h) 250 mg/kg SC 250 mg/kg SC
treatment (0h) bromocriptine bromocriptine
mg/kg SC 10 mg/kg SC
activity count (0.5 - 1.5h) 17 36

This model was used to evaluate whether or not Compound 10 exhibits the same
superiority as L-
DOPA and apomorphine over D2 agonists. A dose response experiment for compound
10 was
5 performed and there was a dose-dependent reversal of the hypomotility
deficits induced by
severe depletion of endogenous dopamine. A final experiment directly comparing
the effects of
apomorphine, pramipexole and compound 10 in this model was performed and
confirmed that
compound 10 was able to restore locomotion in MPTP mice treated and was
superior to
Pramipexole in this model. The data is presented in Table 4e.
Table 4e. Superiority of apomorphine and compound 10 over Pramipexole in the
mouse
MPTP/AMPT model.

AMPT AMPT
pretreatment (-1.5h) vehicle 250 mg/kg SC 250 mg/kg SC
treatment (0h) vehicle vehicle apomorphine
l mg/kg SC
activity count (0.5 -1.5h) 509 2 904
AMPT AMPT
pretreatment (-1.5h) 250 mg/kg SC 250 mg/kg SC
treatment (0h) pramipexole compound 10
1 mg/kg SC 0.030 mg/kg SC
activity count (0.5 - 1.5h) 176 690

Based on the above data in Tables 4a-4e, and in one embodiment of the
invention, it is expected
that compound 10 can be used to treat a 'moderate-to-severe PD' or 'severe PD'
patient
population.

The lower induction of dyskinesias by compound 10 relative to apomorphine and
L-DOPA
combined with the Dl/D2 dissection study (and the MPTP/AMPT mouse + MPTP
marmosets
studies) supports first-line treatment with compound 10. Today, D2 agonists
such as pramipexole


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
59
are preferred first-line medication due to their better 'fluctuation side-
effects' profile (e.g.
dyskinesias) as compared to L-DOPA. Our data demonstrates that compound 10 is
as efficacious
as L-DOPA (and apomorphine) but that it also has a better dyskinesia profile
than L-DOPA and
apomorphine. Since L-DOPA is consistenly more effiacious than D2 agonists like
pramipexole in
all stages of PD, it is believed that compound 10 would be an optimal drug for
first-line treatment
based on the combined dual D1/D2 profile in vivo, efficacy on par with L-DOPA
and better than
D2 agonist, and with a dyskinesia profile better than L-DOPA.

Example 11: Pharmacological Testing in vivo V
Anti-Parkinsonian effects in MPTP-treated common marmosets
The experiments were conducted using 6 MPTP treated marmosets (2.Omg/kg daily
for up to 5
consecutive days dissolved in sterile 0.9% saline solution). All the animals
had previously been
treated with L-DOPA (12.5mg/kg p.o., plus carbidopa 12.5mg/kg p.o.)
administered daily for up
to 30 days in order to induce dyskinesia. Prior to the study all subjects
exhibited stable motor
deficits including a marked reduction of basal locomotor activity, poor
coordination of
movement, abnormal and/or rigid posture, reduced alertness and head checking
movements.
Domperidone was administered 60 min before any of the test compounds.
Domperidone is an
antidopaminergic drug that suppresses nausea and vomiting. Locomotor activity
was assessed
using test cages that are comprised of 8 photo-electric switches comprised of
8 infra-red beams
which are strategically placed in the cage and interruption of a beam is
recorded as one count.
The total number of beam counts per time segment is then plotted as time
course or displayed as
area under the curve (AUC) for total activity. The assessment of motor
disability was performed
by a trained observer blinded to the treatment.

L-DOPA (12.5mg/kg, p.o.) increased locomotor activity and reversed motor
disability as
previously described (Smith et at., Mov. Disord. 2002, 17(5), 887). The dose
chosen for this
challenge is at the top of the dose response curve for this drug. Compound 10
(administered
subcutaneously (0.001 or 0.01 mg/kg SC) produced a dose-related increase in
locomotor activity
and reversal of motor disability tending to produce in a response greater than
for L-DOPA
(12.5mg/kg, p.o.). Compound 10 produced prolonged reversal of motor disability
compared to L-
DOPA and was as efficacious as L-DOPA. This data is presented in Table 5.


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
Table 5. Mean disability scores of MPTP-marmosets when treated with L-DOPA or
compound
10.

group 1 group 2 group 3 group 4
treatment vehicle L-DOPA compound 10 compound 10
12.5 mg/kg PO 0.001 mg/kg SC 0.01 mg/kg SC
disability score 13.0 10.0 14.3 10.0
(60 min)
disability score 11.0 2.0 2.3 2.2
(120 min)
disability score 11.0 1.8 2.5 2.0
(180 min)
disability score 12.7 3.2 3.0 2.2
(240 min)
disability score 12.2 5.0 2.5 2.7
(300 min)
disability score 13.0 9.7 6.5 2.3
(360 min)
disability score 13.3 11.0 8.5 2.7
(420 min)

5 Example 12: Pharmacological Testing in vivo VI
Reversal of reserpine-induced hypomotility by buccal delivery of compound 10
Rats weighing ca. 200g were treated with reserpine (5 mg/kg subcutaneously as
a solution in
20% aqueous solutol for which pH was adjusted to 4 with methanesulfonic acid).
Administering
reserpine to rats depletes presynaptic nerve endings from dopamine and
therefore reserpinesed
10 rats are temporarily `parkinsonian' and unable to move unless treated with
a dopamine agonist or
L-DOPA. A separate group of four animals was treated subcutaneously with the
vehicle used for
reserpine (group 1). After 23-24 hours the 24 reserpine animals were divided
into groups 2-6 with
four animals in each. These were treated as summarized below, before they were
placed in
activity boxes equipped with photosensors and their locomotor activity was
recorded over 3
15 hours. Group 1: treated with 20% ethanol in 0.7% aqueous sodium chloride
subcutaneously.
Group 2: treated with apomorphine (1 mg/kg administered subcutaneously as an
aqueous solution
with pH = 4. 0.02% ascorbic acid had been added to prevent decomposition of
apomorphine).
Group 3: treated with compound 10 (administered subcutaneously as solution in
20% ethanol in
0.7% aqueous sodium chloride). Groups 4-6: treated with increasing doses of
compound 10
20 (administrated buccally in the upper right gingival as a solution in 20%
ethanol in 0.7% aqueous
sodium chloride). The data showed that apomorphine (1 mg/kg subcutaneously;
positive control)
and compound 10 (administered subcutaneously) reversed the reserpine-induced
hypomotility.


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
61
Compound 10 (administered buccally) reversed the hypomotility. The data is
summarized in
Table 6.

Table 6. Effect of apomorphine (dosed subcutaneously) and compound 10 (dosed
buccally) in the Ungerstedt model.

group 1 group 2 group 3
treatment (23-24 h prior reserpine reserpine
to activity measurement) vehicle 5 mg/kg SC 5 mg/kg SC
treatment (Oh prior to apomorphine compound 10
activity measurement) vehicle 1 mg/kg SC 0.01 mg/kg SC
activity count 486 440 308
group 4 group 5 group 6
treatment (23-24 h prior reserpine reserpine reserpine
to activity measurement) 5 mg/kg SC 5 mg/kg SC 5 mg/kg SC
treatment (Oh prior to compound 10 compound 10
activity measurement) vehicle buccally 0.05 mg/kg 0.10 mg/kg
buccally buccally
activity count 17 378 533
Example 13: Pharmacological Testing in vivo VII

Induction of rotation response in 6-OHDA rats by buccal delivery of compound
10

We have used rats with unilateral 6-OHDA lesions to assess compound 10 for its
ability to induce
rotation after buccal administration [for details on the model, see the
description under example
7]. A group of eight animals was treated with apomorphine (positive control;
0.1 mg/kg
administered subcutaneously as an aqueous solution with pH = 4. 0.02% ascorbic
acid had been
added to prevent decomposition of apomorphine). Another two groups of eight
animals were
treated with two different doses of compound 10 (administered buccally in the
upper right
gingiva as a solution in 20% ethanol in 0.7% aqueous sodium chloride).
Apomorphine induced
rotations after subcutaneous administration. Buccal delivery of compound 10
also induced
circling behavior. The data is summarized in Table 7.


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
62
Table 7. Effect of apomorphine (dosed subcutaneously) and compound 10 (dosed
buccally) in the Ungerstedt model.

apomorphine compound 10 compound 10
dose 0.1 mg/kg SC 0.01 mg/kg 0.1 mg/kg
buccally buccally
mean number of 1123 910 1203
rotations over 3h

Example 14: Pharmacological Testing in vivo VIII

Intravenous and Buccal pharmacokinetic study in the Minipig
The objective of this study was to determine the plasma concentrations of
compound 10 in
minipig following dosing with compound 10 (by either intravenous
administration at 0.0025
mg/kg or by buccal administration at 0.0 10 mg/kg and 0.040 mg/kg).

STUDY DESIGN
Test and control articles
The test article was compound 10. The vehicles for the test article were
Sterile saline (0.9%
NaCl) (intravenous administration) supplied by Baxter, Norfolk or Ascorbic
acid reconstituted in
Water for Injection (buccal administration) supplied by VWR International,
Leicestershire.
Formulations were prepared on the day of dosing.

Test system and dose levels
Three male minipigs of the Gottingen ApS strain were supplied by Ellegaard
Gottingen,
Dalmose, Denmark. At initiation of dosing, animals were approximately 15 to 17
weeks old.
Each animal was dosed once on three separate occasions according to the
following study design:

G oly f T0111p Dose level D >e ,s. Duane OccasiEa Route _'`nir21 ti.:i:iber s
in untie! lescitp s.na tEi'?lc~? (Study Da i :tale

1 Lcra.- ? 5 L1 tanL:'1
1iõi'azen su> B':Ahisl 1-3
I
1 Low 1+:i 10 t:_L: l ? ; :c a 1-3
1 F is 2 40 10 t L 1_g 5 i 33eti.a 1-31

Animals were deprived of food overnight and anaesthetised with isoflurane in
oxygen
(administered by facemask), prior to each dosing occasion.


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
63
Day 1- Intravenous administration
Intravenous administrations were performed by slow manual injection via a
temporary catheter
placed in the ear vein whilst under anaesthesia, animals were allowed to
recover from the
anaesthesia immediately after dosing. Whilst anaesthetised, a catheter was
inserted into the
jugular vein and secured in place for the purpose of blood collection. The
catheter was filled with
heparin (250 iu/mL in 0.9% sodium chloride). The exterior portion of the
catheter was routed
from the ventral neck to the dorsum of the minipig and protected by bandaging.
The distal end of
the catheter was capped and placed in a re-sealable pouch within the bandage.
The jugular
catheter was retained in place and flushed with heparinised saline every 24
hours.
Days 3 and 5 - Buccal administrations
Buccal administrations were performed by applying the test formulation to the
buccal membrane
for 5 minutes while the animal was anaesthetised. Any residual formulation
remaining in the
mouth after the 5 minute application was left in the mouth. Animals were
allowed to recover
from the anaesthesia immediately after dosing.

Plasma Concentrations
Blood samples were taken from all animals on Day 1 following intravenous
(bolus)
administration, all animals on Day 3 following buccal administration of a low
dose and all
animals on Day 5 following buccal administration of a high dose for
pharmacokinetic analysis.
The samples (1.0 mL) were collected from the jugular vein (via catheter) into
tubes containing
EDTA anticoagulant. Prior to addition of the blood sample, 100 microL of a
stabiliser (2% beta-
mercaptoethanol containing 20 mg/mL ascorbic acid) was added to each pot. The
stabiliser was
prepared fresh on each day of sample collection. Samples were collected as
follows:
= Day 1: 5, 10, 15, 30 and 45 minutes and 1, 2, 4, 6, 8, 12 and 16 hours post-
dose
= Day 3: pre-dose and at 5, 10, 15, 30 and 45 minutes and 1, 2, 4, 6, 8, 12,
16 and 24
hours post-dose
= Day 5: pre-dose and at 5, 10, 15, 30 and 45 minutes and 1, 2, 4, 6, 8, 12,
16 and 24
hours post-dose
The times of the blood sampling were generally adhered to. The greatest
deviation from the
scheduled timepoints was one minute late at the 5 minute timepoint on Day 3.
The blood samples
were centrifuged within one hour of sample collection and the resultant plasma
was frozen prior
to analysis.


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
64
Sample Preparation Procedure

Step Procedure
1 Thaw frozen quality control samples, control matrix and matrix study
samples and calibration standards at room temperature.
2 Vortex mix samples (ca. 10 seconds).
3 Centrifuge (ca. 10 minutes, ca. 3500 rpm, room temperature) in the bench
top centrifuge or corresponding 'g' force in a micro-centrifuge.
4 Aliquot calibration standards, QCs, study samples and blanks (100 LL)*
into a 2 mL 96 deep well plate.
Return unused portion of samples to freezer.
Add internal standard solution (500 L, solution IS C) using a repeating
6 pipette, to all wells except blanks, which receive 500 gL of 100 mM
ammonium formate (aq) + 1% formic acid.
7 Cap the plate and gently mix on a plate mixer (ca. 5 minutes).

8 Centrifuge the plate in a bench top centrifuge (ca. 3500 rpm, 10 minutes,
room temperature).
9 Prime SPE plate (Oasis HLB 10 mg) with methanol (500 L per well). Use
minimum pressure or gravity and do not allow to dry.
Prime plate with water (500 L per well) using minimum pressure.
11 Transfer samples (approximately 500 L) to plate using an automatic 8
channel pipette.
12 Pass through plate using minimum pressure.
13 Wash plate with water: methanol (90:10 v/v) (500 L) using minimum
pressure and then increase pressure to maximum for one minute.
Slowly elute sample into 1.2 mL 96 deep well plate with 20 mM
14 ammonium formate (aq): acetonitrile: formic acid (50:50:2 v/v/v) (250 L)
using minimum pressure and then increase pressure to dry packing material
completely.
Pulse spin the plate containing the eluate to 1000 rpm in a bench-top
centrifuge (place into centrifuge, spin up to 1000 rpm and then stop).
Evaporate the acetonitrile composition of the eluate under a stream of
16 nitrogen (nominal30 C) for a minimum of 30 minutes and until an
estimated half of the original volume remains
17 Add 100 L of (20 mM ammonium formate (aq) + 0.5% formic acid):
acetonitrile (90:10 v/v) containing 4 mg/mL ascorbic acid to each well.
18 Cap the plate and vortex mix (ca. 2 minutes).
19 Centrifuge the plate in a bench top centrifuge (ca. 3500 rpm, 10 minutes,
nominal room temperature).
Submit for analysis.


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
Analytical Methods
The plasma concentrations of compound 10 were determined after solid phase
extraction of the
plasma samples followed by high performance liquid chromatography with tandem
mass
spectrometric detection (LC-MS/MS) using a sample volume of about 100 microL.
5
Internal standard solution, containing the internal standard of compound 10
was added to thawed
plasma samples (100 microL aliquot). The SPE plate (Oasis HLB, 10 mg) was
conditioned with
methanol (500 microL) followed by water (500 microL). The sample (approx. 500
microL
aliquot) was transferred to the pre-conditioned SPE plate. The sample was then
passed through
10 the cartridge, which was then washed with water: methanol (90: 10 v/v, 0.5
mL). The sample was
then eluted into a fresh 96 well polypropylene collection plate with 20 mM
ammonium formate
(aq): acetonitrile: formic acid (50: 50: 2 v/v/v, 250 microL). The organic
component of the eluted
samples was then evaporated under a gentle stream of nitrogen until
approximately 50% of the
original volume was remaining. An aliquot (100 microL) of a solution
containing 20 mM
15 ammonium formate (aq) and 0.5% formic acid: acetonitrile (90: 10 v/v)
together with 4 mg/mL
ascorbic acid was added to the remaining aqueous component of the sample in
each well, vortex
mixed, centrifuged (3500 rpm, 10 minutes, room temperature) prior to being
submitted for
UHPLC-MS/MS analysis.

20 Concentrations of compound 10 in calibration standards, QC samples and
study samples were
determined using least squares linear regression with 1/x weighting for
compound 10. The
plasma concentrations of compound 10 were determined after solid phase
extraction of the
plasma samples followed by high performance liquid chromatography with tandem
mass
spectrometric detection (LC-MS/MS). The method was validated and has a lower
limit of
25 quantification (LLOQ) of 10 pg/mL using 100 microL of plasma.

Analytical Procedure: Liquid chromatography - tandem mass spectrometry (LC-
MS/MS) API
5000: Final extract solutions were submitted for LC-MS/MS analysis under the
following
conditions.
LC conditions:
Analytical column# Waters BEH UPLC Phenyl 100 x 2.1 mm column,
1.7 microm particle size, part number 186002885
In line filter (Acquity) Supplier: Waters Part n/o 700002775


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
66
Column oven temperature# Nominal 50'C
Autosampler temperature Nominal 4 C
Mobile phase A# 20 mM ammonium formate (aq) + 0.5 % formic acid
Mobile phase B# Acetonitrile
Flow rate# 0.5 mL/min
Gradient settings: See table below

Time kS'x1:tLi:Tf':

6_0 5
`.1 35 y
9.0 3 y
Switching valve times 0 - 1.2 mins - To waste
10 1.2-6mins-ToMS
6 - 8 mins - To waste
Slave pump solvent (20 mM ammonium formate (aq)+ 0.5% formic acid):
acetonitrile (50:50 v/v)
Slave pump flow rate 0.5 mL/min
15 Wash solvent 1# (20 mM ammonium formate (aq)+ 0.5% formic acid):
Weak wash (Acquity) acetonitrile (90:10 v/v)
Wash solvent 2# Water: methanol: TFA (50:50:0.1 v/v/v)
Strong wash (Acquity)
Injection mode (Acquity) partial loop with needle over-fill
20 LC conditions
Injection loop volume (Acquity) 50 microL
Needle placement 2.0 mm from bottom
Injection volume (Recommended) 50 microL
Waters acquity
25 Weak wash volume ( L) 3000 (Range 200 to 5000)
Strong wash volume ( L) 3000 (Range 0 to 5000)
Mass spectrometer parameters API 5000
Mode of operation# Turbo IonSpray (Positive ion) (MS/MS)


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
67
Collision gas setting (CAD) 6 [Where a setting of 12 is approximately equal to
4.8 x
10-5 Torr for a API 4000 instrument]
Curtain gas setting (CUR) 20 psi
Ion source gas 1 (GS1) 50 psi
Ion source gas 2 (GS2) 70 psi
IonSpray voltage (IS) 5500 V
Temperature (TEM) 650 C
Q1 Resolution Unit
Q3 Resolution Low
Interface heater status On
Analysis time 6 minutes in two periods:
Period one: 3.5 minutes
Period two: 2.5 minutes

A representative chromatogram generated using the above procedure and acquired
during the
determination of compound 10 in minipig plasma is presented in Figure 3. As
the quantification
of compound 10 was based upon peak height ratios, the integrations on some of
the
chromatograms include additional noise and interference peaks to ensure the
correct peak height
is measured.
Plasma concentrations of compound 10 following intravenous administration
Plasma concentrations for compound 10 following single intravenous bolus
administration of
compound 10 at 0.0025 mg/kg. The data are summarized in Table 8.


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
68
Table 8. Plasma concentrations of compound 10 in minipigs following
intravenous
administration of compound 10 (0.0025 mg/kg)

Animal Day I Day I Day I Day I Day I Day I Day I
Hour 0.08 Hour 0.17 Hour 0.25 Hour 0.5 Hour 0.75 Hour 1 Hour 2
1 1920 1260 828 536 667 451 198
2 1210 834 740 562 534 379 196
3 1720 976 1170 922 656 432 352
Mean
1620 1020 913 673 619 421 249
(pg/ml)
SD (n-1) 366 217 227 216 73.8 37.3 89.5
Animal Day I Day I Day I Day I Day I
Hour 4 Hour 6 Hour 8 Hour 12 Hour 16
1 74.8 47.6 24.5 17.0 8.58
2 76.3 56.2 33.9 29.9 9.25
3 115 75.6 40.7 20.9 10.1
Mean
88.7 59.8 33.0 22.6 9.31
(pg/ml)
SD (n 1) 22.8 14.3 8.13 6.62 0.762
Following single intravenous bolus administration of compound 10 at 0.0025
mg/kg to male
minipigs, maximum plasma concentrations of compound 10 were observed at 5
minutes post-
dose, i.e. at the first blood sampling time post intravenous administration.
Plasma concentrations
of compound 10 appeared to decline in a generally bi-phasic manner with an
apparent terminal
elimination half-life (tl/2) ranging from 3.4 to 4.3 hours, with the start of
the apparent terminal
phase occurring at 4 hours post-dose.

Over the 16 hour sampling period, plasma concentrations were quantifiable
(i.e. above the LLOQ
of 10 pg/mL) up to 12 hours post-dose in 2 animals, with concentrations
estimated at 16 hour
post-dose as levels were above 20% of the LLOQ. In one animal (animal 3),
plasma
concentrations were greater than the LLOQ throughout the 16 hour period.


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
69
Plasma concentrations of compound 10 following buccal administration
Plasma concentrations of compound 10 in minipigs following buccal
administration (0.010
mg/kg). The data are summarized in Table 9.

Table 9. Plasma concentrations of compound 10 in minipigs following buccal
administration of compound 10 (0.010 mg/kg)

Animal Day 3 Day 3 Day 3 Day 3 Day 3 Day 3 Day 3
Hour 0.08 Hour 0.17 Hour 0.25 Hour 0.5 Hour 0.75 Hour 1 Hour 2
1 379 105 185 758 1100 791 501
2 55.2 221 622 556 964 817 372
3 16.1 117 134 943 1070 1000 589
Mean
150 148 314 752 1040 869 487
(pg/mi)
SD (n-1) 199 63.8 268 194 71.5 114 109
Day I Day I Day I Day I Day I Day I
Animal Hour Hour 24
Hour 4 Hour 6 Hour 8 Hour 16
12
1 98.3 84.5 46.7 26.8 6.91 <2.00
2 91.7 26.5 26.1 23.4 NR 4.94
3 206 75.8 55.4 157 NS 26.1
Mean
132 62.3 42.7 69.1 6.91 15.5
(pg/mi)
SD (n-1) 64.2 31.3 15.0 76.2
NR: No result reported
NS: No Sample

Plasma concentrations of compound 10 in minipigs following buccal
administration (0.040
mg/kg). The data are summarized in Table 10.


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
Table 10. Plasma concentrations of compound 10 in minipigs following buccal
administration of compound 10 (0.040 mg/kg).

Animal Day 5 Day 5 Day 5 Day 5 Day 5 Day 5 Day 5
Hour 0.08 Hour 0.17 Hour 0.25 Hour 0.5 Hour 0.75 Hour 1 Hour 2
1 695 1930 3330 8230 10700 10800 6340
2 427 1820 3890 5310 10500 9580 3880
3 2750 6060 9140 7880 15900 11900 8680
Mean
1290 3270 5450 7140 12400 10760 6300
(pg/ml)
SD (n-1) 1270 2420 3210 1590 3060 1160 2400
Day I Day I Day I Day I Day I Day I
Animal Hour Hour 24
Hour 4 Hour 6 Hour 8 Hour 16
12
1 2560 781 445 272 142 44.3
2 746 278 212 139 58.4 78.9
3 2490 1730 910 1400 677 233
Mean
1930 930 522 604 292 119
(pg/ml)
SD (n-1) 1030 737 355 693 336 100
5
Following single buccal administration of compound 10 at 0.010 mg/kg and 0.040
mg/kg to the
male minipig, compound 10 was rapidly absorbed, with compound 10 being
quantifiable in
plasma at 5 minute post-dose. Maximum plasma concentrations were observed at
about 0.75
hours post-dose, with the exception of animal 1 at the 0.040 mg/kg dose level
with a delayed
10 tmax of 1 hour post-dose. After attainment of Cmax, plasma concentrations
of compound 10
appeared to decline in a bi-phasic manner, with mean apparent terminal half-
lives of 5.1 and 5.6
hours at the 0.010 mg/kg and 0.040 mg/kg dose levels, respectively.

Over the 24 hour sampling period, plasma levels of compound 10 remained above
the LLOQ,
15 apart from 2 animals following the 0.010 mg/kg dose where plasma
concentrations were either


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
71
estimated (as levels were above 20% of the LLOQ; 16 hour post-dose for 1M; 24
hour post-dose
for 2M), or were not quantifiable (being < 20% of LLOQ; 24 h post-dose for
1M).

Dose proportionality
Fold increases in systemic exposure to compound 10 following increases in dose
from 0.010
mg/kg and 0.040 mg/kg compound 10 are presented below.

D :se

Da-',es ncteem t N -A, 4.. 0
it AUK:
~_~.
U`C, NA 3.tf
9
Lie e az.e tit tea:; NA

Systemic exposure to compound 10 increased in a supra-proportional manner over
the 0.010
mg/kg and 0.040 mg/kg dose range with AUG. and C. increasing by 12-fold over
the 4-fold
increase in dose. The bioavailability of compound 10 following buccal
administration was dose
dependent, ranging from 30 to 42% at 0.010 mg/kg, increasing to 73 to 136% at
0.040 mg/kg
The dose normalised AUCO-c and Cmax for compound 10 are presented graphically
in Figures
3 and 4, respectively.

Conclusion
Following intravenous bolus administration of 0.025 mg/kg compound 10 to male
minipigs,
plasma concentrations of compound 10 appeared to decline in a bi-phasic manner
with individual
apparent terminal elimination half-life ranging from 3.4 to 4.3 hours.

Absorption of compound 10 was rapid following single buccal administration of
compound 10,
with maximum plasma concentrations being observed at 0.75 to 1 hours post-
dose. Plasma
concentrations of compound 10 appeared to decline in a bi-phasic manner and
the apparent
terminal elimination half-life was independent of dose, with values ranging
from 3.1 to 5.6 hours
in individual animals.
Following buccal administration, systemic exposure to compound 10 appeared to
increase in a
supra-proportional manner with a 12-fold increase in both AUCO-00 and Cmax
over the 0.010 to
0.040 mg/kg dose range. Due to the non-linearity in exposure, bioavailability
of compound 10


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
72
was dose dependent with mean values of 31 to 35% at 0.010 mg/kg increasing to
105 to 122% at
0.040 mg/kg.

Example 15: Pharmacological Testing in vivo IX
Induction of circling behaviour in a rat model of Parkinson's disease by
intranasal administration of
compound 10.

Animals were generated as described under example 7. Four groups of animals
were dosed with
various doses of compound 10 (group 1, 1 microg/kg; group 2, 10 microg/kg;
group 3, 25
microg/kg; group 4, 50 microg/kg). In all cases, compound 10 was administered
in one of the
nostrils in a volume of 20 microL of a solution of the appropriate
concentration in 20% ethanol in
0.7% aqueous sodium chloride containing 0.02% ascorbic acid. The drug solution
was applied to
one of the nostrils and the nose was gently massaged to ensure distribution of
the administered
solution over the nasal mucosa. The degree of rotation behaviour of the
animals was recorded over
the next 3 hours. The data are presented in Table 11.

Table 11. Rotation response of unilaterally lesioned 6-OHDA rats over 3 hours
following
intranasal administration of compound 10.

group 1 group 2 group 3 group 4
mean number of 401 691 1286 2122
rotations (0 - 3h)

Example 16: Pharmacological Testing in vivo X

Induction of rotation response in 6-OHDA rats by transdermal delivery of
compound 10

We have used rats with unilateral 6-OHDA lesions to assess compound 10 for its
ability to induce
rotation after transdermal administration [for details on the model, see the
description under
example 7]. Three groups of six animals were treated with different doses of
compound 10
administered transdermally. Compound 10 (24 mg) was suspended in a mixture of
0.02% ascorbic
acid and 20% ethanol in saline (9 mL); the resulting suspension was diluted
with dimethyl
sulfoxide (0.45 mL). The appropriate amount of this formulation was applied to
the ears of the
animals. The ears were rubbed gently before the rotation response of the
animals was assessed over
3h. Transdermal delivery of compound 10 induced circling behavior in all three
groups. The data is
summarized are Table 12.


CA 02751321 2011-08-02
WO 2010/097091 PCT/DK2010/050050
73
Table 12. Effect of compound 10 (dosed transdermally) in the Ungerstedt model.

group 1 group 2 group 3
0.127 mg/kg 0.254 mg/kg 0.381 mg/kg
dose compound 10 compound 10 compound 10
transdermally transdermally transdermally
mean number of 482 837 1448
rotations over 3h

Representative Drawing

Sorry, the representative drawing for patent document number 2751321 was not found.

Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date Unavailable
(86) PCT Filing Date 2010-02-26
(87) PCT Publication Date 2010-09-02
(85) National Entry 2011-08-02
Dead Application 2015-02-26

Abandonment History

Abandonment Date Reason Reinstatement Date
2014-02-26 FAILURE TO PAY APPLICATION MAINTENANCE FEE

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee $400.00 2011-08-02
Maintenance Fee - Application - New Act 2 2012-02-27 $100.00 2012-02-14
Maintenance Fee - Application - New Act 3 2013-02-26 $100.00 2013-02-06
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
H. LUNDBECK A/S
Past Owners on Record
None
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Abstract 2011-08-02 1 66
Claims 2011-08-02 3 87
Drawings 2011-08-02 5 69
Description 2011-08-02 73 3,680
Cover Page 2011-09-23 2 43
PCT 2011-08-02 1 48
Assignment 2011-08-02 5 161
Correspondence 2011-09-16 1 81
Correspondence 2011-10-27 1 50